Skip to main content

Full text of "USPTO Patents Application 10551504"

See other formats


(12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) 



(19) World Intellectual Property Organization 

International Bureau 

(43) International Publication Date 
27 December 2001 (27.12.2001) 




(10) International Publication Number 

PCT WO 01/97858 A2 



(51) International Patent Classification 7 : A61K 51/00 (81) Designated States (national): AE, AG, AL, AM, AT, AU, 



(21) International Application Number: PCT/US0 1/1 8939 

(22) International Filing Date: 14 June 2001 (14.06.2001) 
(25) Filing Language: English 



(26) Publication Language: 

(30) Priority Data: 

60/212,668 



20 June 2000 (20.06.2000) US 



(71) Applicant: IDEC PHARMACEUTICALS CORPORA- 
TION [US/US]; 3030 Callan Road, San Diego, CA 92121 
(US). 

(72) Inventor: WHITE, Christine; P.O. Box 9242, Rancho 
Santa Fe, CA 92067 (US). 

(74) Agents: TESKIN, Robin, L. et ai.; Pillsbury Winthrop 
LLP, 1600 Tysons Boulevard, McLean, VA 22102 (US). 



AZ, BA, BB, BG, BR, BY, BZ, CA, CH, CN, CO, CR, CU, 
CZ, DE, DK, DM, DZ, EC, EE, ES, Fl, GB, GD, GE, GH, 
GM, HR, HU, ID, IL, IN, IS, JP, KE, KG, KP, KR, KZ, LC, 
LK, LR, LS, LT, LU, LV, MA, MD, MG, MK, MN, MW, 
MX, MZ, NO, NZ, PL, PT, RO, RU, SD, SE, SG, SI, SK, 
SL, TJ, TM, TR, TT, TZ, UA, UG, UZ, VN, YU, ZA, ZW. 



English Designated States (regional): ARIPO patent (GH, GM, 

KE, LS, MW, MZ, SD, SL, SZ, TZ, UG, ZW), Eurasian 
patent (AM, AZ, BY, KG, KZ, MD, RU, TJ, TM), European 
patent (AT, BE, CH, CY, DE, DK, ES, Fl, FR, GB, GR, IE, 
IT, LU, MC, NL, PT, SE, TR), OAPI patent (BF, BJ, CF, 
CG, CI, CM, GA, GN, GW, ML, MR, NE, SN, TD, TG). 



Published: 

— without international search report and to be republished 
upon receipt of that report 

For two-letter codes and other abbreviations, refer to the "Guid- 
ance Notes on Codes and Abbreviations" appearing at the begin- 
ning of each regular issue of the PCT Gazette. 



< 

id — ■ 

00 (54) Title: TREATMENT OF B-CELL ASSOCIATED DISEASES SUCH AS MALIGNANCIES AND AUTOIMMUNE DIS- 

^ EASES USING A COLD ANTI-CD20 ANTIBODY/RADIOLABELED ANTI-CD22 ANTIBODY COMBINATION 
ON 

(57) Abstract: Treatment of B-cell associated diseases including autoimmune and B-cell malignancies such as leukemias, lym- 
phomas, using the combination of an anli-CD20 antibody, preferably RITUXAN® and a radiolabeled anti-CD22 antibody, prefer- 

Q ably an "Y labeled humanized anti-CD22 antibody, is described. These therapeutic regimens provide for enhanced depletion of B 
cells, and therefore reduce the risk in B cell malignancy treatment of relapse associated with RITUXAN® and, moreover, provide 

^ for prolonged immunosuppression of B-cell immune responses, especially in the context of autoimmune diseases and transplant 



WO 01/97858 



PCT/US01/18939 



TREATMENT OF B-CELL ASSOCIATED DISEASES SUCH AS 
MALIGNANCIES AND AUTOIMMUNE DISEASES USING A COLD 
ANTI-CD20 ANTD3QDY/RADIOLABELED ANTI-CD22 
ANTIBODY COMBINATION 

5 

Cross Reference to Related Application 

This application claims priority from U.S. Provisional Serial No. 60/212,668, 
filed June 20, 2000, and which is incorporated herein in its entirety by reference. 

10 Field of the Invention 

The present invention is concerned with a combination immunotherapy/- 
radiotherapy involving the administration of a cold anti-CD20 antibody, preferably 
RITUXAN®, or another anti-CD20 antibody having substantially the same B-cell 
depleting activity as RITUXAN®, and a radiolabeled anti-CD22 antibody, preferably 
15 an yttrium labeled humanized anti-CD22 antibody. In the case of tumor therapy, the 
initial administration of the cold anti-CD23 antibody helps remove B cells from the 
circulation, thereby improving the targeting and efficacy of the radiolabeled anti- 
CD22 antibody. 

Also, the subject treatment provides for enhanced immunosuppression vis-a- 
20 vis cold CD20 and radiolabeled anti-CD22 therapy alone. This combination 

therapeutic regimen is useful in the treatment of diseases wherein depletion and/or 
selective killing, and/or blocking the function of CD20 and CD22 expressing cells is 
therapeutically beneficial, especially B-cell malignancies, lymphomas, leukemias, and 
conditions or diseases wherein suppression of B-cell immune function is . 
25 therapeutically beneficial, e.g., autoimmune diseases, allergic diseases, transplant, and 
other therapeutic regimens involving administration of antigenic moieties, e.g., 

y 

protein, cell or gene therapy. Preferably, the therapeutic regimen will comprise the 
initial administration of RITUXAN®,' followed by admhiistration of the radiolabeled 
anti-CD22 antibody. 

30 

Background of the Invention 
I. Anti-CD20 Antibodies 

CD20 is a cell surface antigen expressed on more than 90% of B-cell 
lymphomas and does not shed or modulate in the neoplastic cells (McLaughlin et al., 



WO 01/97858 



PCT/US01/18939 



J. Clin. Oncol. 16: 2825-2833 (1998)). Anti-CD20 antibodies have been prepared for 
use both in research and therapeutics. One reported anti-CD20 antibody is the 
monoclonal Bl antibody (U.S. Patent No. 5,843,398). Anti-CD20 antibodies have 
also been prepared in the form of radionuclides for treating B-cell lymphoma (e.g. , 
5 131 I-labeled anti-CD20 antibody), as well as a 89 Sr-labeled form for the palliation of 
bone pain caused by prostate and breast cancer metastasises (Endo, Gan To Kagaku 
Ryoho 26: 744-748 (1999)). 

A murine monoclonal antibody, 1F5, (an anti-CD20 antibody) was reportedly 
adrninistered by continuous intravenous infusion to B cell lymphoma patients. 

10 However, extremely high levels (>2 grams) of 1F5 were reportedly required to deplete 
circulating tumor cells, and the results were described as "transient" (Press et at, 
Blood 69: 584-591 (1987)). A potential problem with using monoclonal antibodies 
as therapeutics is that non-human monoclonal antibodies (e.g., murine monoclonal 
antibodies) typically lack human effector functionality, e.g., they are unable to, inter 

1 5 alia, mediate complement dependent lysis or lyse human target cells through 
antibody-dependent cellular toxicity or Fc-receptor mediated phagocytosis. 
Furthermore, non-human monoclonal antibodies can be recognized by the human host 
as a foreign protein; therefore, repeated injections of such foreign antibodies can lead 
to the induction of immune responses leading to harmful hypersensitivity reactions. 

20 For murine-based monoclonal antibodies, this is often referred to as a Human 

Anti-Mouse Antibody response, or "HAMA" response. Additionally, these "foreign" 
antibodies can be attacked by the immune system of the host such that they are, in 
effect, neutralized before they reach their target site. 
A. RITUXIMAB® 

25 RITUXIMAB® (also known as RITUXAN®, MabThera® and IDEC-C2B8) 

was the first FDA-approved monoclonal antibody and was developed at DEC 
Pharmaceuticals (see U.S. Patent Nos. 5,843,439; 5,776,456 and 5,736,137). 
RITUXIMAB® is a chimeric, anti-CD20 monoclonal (MAb) recommended for 
treatment of patients with low-grade or follicular B-cell non-Hodgkin's lymphoma 

30 (Mclaughlin et al, Oncology (Huntingt) 12: 1763-1777 (1998); Leget et al, Curr. 
Opin. Oncol. 10: 548-551 (1998)). m Europe, RITUXIMAB® has been approved for 
therapy of relapsed stage IMV follicular lymphoma (White et al, Pharm. Sci. 



2 



WO 01/97858 



PCT/US01/18939 



Technol. Today 2: 95-101 (1999)). Other disorders treatable with. RTTUXMAB® 
include follicular centre cell lymphoma (FCC), mantle cell lymphoma (MCL), diffuse 
large cell lymphoma (DLCL), and small lymphocytic lymphoma/chronic lymphocytic 
leukemia (SLL/CLL) (Nguyen et al, 1999)). RITUXMAB® has exhibited minimal 
5 toxicity and significant therapeutic activity in low-grade non-Hodgkin's lymphomas 
(NHL) in phase I and II clinical studies (Berinstein et al.,Ann. Oncol. 9: 995-1001 
(1998)). 

RITUXMAB®, which is currently being used alone to treat B-cell NHL at 
weekly doses of typically 375 mg/M 2 for four weeks with relapsed or refractory low- 

1 0 grade or follicular NHL. This antibody is well tolerated and had significant clinical 
activity (Piro etal.,Ann. Oncol. 10: 655-61 (1999); Nguyen et al, Eur. J. Haematol 
62: 76-82 (1999); and Coiffier et al, Blood 92: 1927-1932 (1998)). Also, up to 500 
mg/M 2 of four weekly doses have also been administered during trials using the 
antibody (Maloney et al, Blood 90: 2188-2195 (1997)). RITUXIMAB® also has 

1 5 been combined with chemotherapeutics, such as CHOP (e.g. , cyclophosphamide, 

doxorubicin, vincristine and prednisone), to treat patients with low-grade or follicular 
B-cell non-Hodgkin's lymphoma (Czuczman et al, J. Clin. Oncol. 17: 268-76 (1999); 
and McLaughlin et al, Oncology (Huntingt) 12: 1763-1777 (1998)). However, it has 
not previously been utilized in combination with other therapeutic antibodies. 

20 The synthesis of monoclonal antibodies against CD22 and their use in 

therapeutic regimens has also been reported. CD22 is a B-cell-specific molecule 
involved in B-cell adhesion that may function in homotypic or heterotypic interactions 
(Stamenkovic et al, Nature 344:74 (1990); Wilson et al, J. Exp. Med. 173:137 (1991); 
Stamenkovic et al, Cell 66:1 133 (1991)). The CD22 protein is expressed in the 

25 cytoplasm of progenitor B and pre-B-cells (Dorken et al, J. Immunol. 136:4470 
(1986); Dorken et al, "Expression of cytoplasmic CD22 in B-cell ontogeny. In 
Leukocyte Typing m, White Cell Differentiation Antigens. McMichael et al, eds., 
Oxford University Press, Oxford, p. 474 (1987); Schwarting et al, Blood 65:974 
(1985); Mason et al, Blood 69:836 (1987)), but is found only on the surface of mature 

30 B-cells, being present at the same time as surface IgD (Dorken et al, J. Immunol. 
136:4470 (1986)). CD22 expression increases following activation and disappears 
with further differentiation (Wilson et al, J. Exp. Med. 1 73 : 1 37 (1 99 1); Dorken et al, 



3 



WO 01/97858 



PCT/US01/18939 



J. Immunol. 136:4470 (1986)). In lymphoid tissues, CD22 is expressed by follicular 
mantle and marginal zone B-cells but only weakly by genninal center B-cells (Dorken 
et al, Immunol. 136:4470 (1986); Ling et al, "B-cell and plasma antigens: new and 
previously defined clusters" In Leukocyte Typing HI. White Cell Differentiation 
5 Antigens, McMichael et al, eds., Oxford University Press, Oxford, p. 302 (1987)). 
However, in situ hybridization reveals the strongest expression of CD22 mRNA 
within the germinal center and weaker expression within the mantle zone (Wilson et 
d,J.Exp. Med. 173:137 (1991)). CD22 is speculated to be involved in the regulation 
of B-cell activation since the binding of CD22 mAb to B-cells in vitro has been found 

10 to augment both the increase in intracellular free calcium and the proliferation induced 
after cross-linking of surface Ig (Pezzutto et al, J. Immunol. 138:98 (1987); Pezzutto 
et al, J. Immunol. 140:1791 (1988)). Other studies have determined, however, that the 
augmentation of anti-Ig induced proliferation is modest (Dorken et al, J. Immunol. 
1 36:4470 (1986)). CD22 is constitutively phosphorylated, but the level of 

15 phosphorylation is augmented after treatment of cells with PMA (Boue et al, J. 

Immunol. 140:192(1988)). Furthermore, a soluble form of CD22 inhibits the CD3- 
mediated activation of human T-cells, suggesting CD22 may be important in T-cell - 
B-cell interactions (Stamenkovic et al, Cell 66:1 133 (1991)). 

Ligands that specifically bind the CD22 receptor have been reported to have 

20 potential application in the treatment of various diseases, especially B-cell lymphomas 
and autoimmune diseases. In particular, the use of labeled and non-labeled anti-CD22 
antibodies for treatment of such diseases has been reported. 

For example, Tedder et al, U.S. patent 5,484,892, that purportedly bind CD22 
with high affinity and block the interaction of CD22 with other ligands. These 

25 monoclonal antibodies are disclosed to be useful in treating autoimmune diseases 
such as glomerulonephritis, Goodpasture's syndrome, necrotizing vasculitis, 
lymphadenitis, periarteritis nodosa, systemic lupus erythematosis, arthritis, 
thrombocytopenia purpura, agranulocytosis, autoimmune hemolytic anemias, and for 
inhibiting immune reactions against foreign antigens such as fetal antigens during 

30 pregnancy, myasthenia gravis, insulin-resistant diabetes, Graves' disease and allergic 
responses. 



4 



WO 01/97858 



PCT/US01/18939 



Also, Leung et al, U. S. Patent 5,789,557, disclose chimeric and humanized 
anti-CD22 monoclonal antibodies produced by CDR grafting and tbe use thereof in 
conjugated and unconjugated form for therapy and diagnosis of B-cell lymphomas and 
leukemias. The reference discloses especially such antibodies conjugated to cytotoxic 
5 agents, such as chemotherapeutic drugs, toxins, heavy metals and radionuclides. (See 
U.S! Patent 5,789,554, issued August 4, 1998, to Leung et al, and assigned to 
Immunomedics.) 

Further, PCT applications WO 98/42378, WO 00/20864, and WO 98/41641 
describe monoclonal antibodies, conjugates and fragments specific to GD22 and 

1 0 therapeutic use thereof, especially for treating B-cell related diseases. 

Also, the use of anti-CD22 antibodies for treatment of autoimmune diseases 
and cancer has been suggested. See, e.g., U.S. Patent 5,443,953, issued August 22, 
1995 to Hansen et al and assigned to Immunomedics Inc. that purports to describe 
anti-CD22 immunoconjugates for diagnosis and therapy, especially for treatment of 

15 viral and bacterial infectious diseases, cardiovascular disease, autoimmune diseases, 
and cancer, and U.S. Patent 5,484,892, issued January 16, 1998 to Tedder et al and 
assigned to Dana-Farber Cancer institute, Inc. that purports to describe various 
monoclonal antibodies directed against CD22, for treatment of diseases wherein 
retardation or blocking of CD22 adhesive function is therapeutically beneficial, 

20 particularly autoimmune diseases.) These references suggest that an anti-CD22 
antibody of fragment may be directly or indirectly conjugated to a desired effector 
moiety, e.g., a label that may be detected, such as an enzyme, fluorophore, 
radionuclide, electron transfer agent during an in vitro immunoassay or in vivo 
imaging, or a therapeutic effector moiety, e.g., a toxin, drug or radioisotope. 

25 Further, an anti-human CD22 monoclonal antibody of the IgGl isotype is 

commercially available from Leinco Technologies, and reportedly is useful for 
treatment of B-cell lymphomas and leukemias, including hairy cell leukemia. 
(Campana, D. et al, J. Immunol. 134:1524 (1985)). Still further, Dorken et al, J. 
Immunol. 150:4719 (1993) and Engel et al, J. Immunol. 150:4519 (1993) both 

30 describe monoclonal antibodies specific to CD22. 

Also, the combined administration of an anti-CD22 immunotoxin and an anti- 
CD 1 9 immunotoxin has been reported for the treatment of diseases including cancer 



5 



WO 01/97858 



PCT/US01/18939 



and autoimmune diseases. (See U.S. Patent 5,686,072, issued November 11, 1997 to 
Uhr et al land assigned to The University of Texas.) 

Therefore, based on the foregoing, while RTTUXAN® and other therapies 
have been reported for treatment of B-cell lymphomas, often such treatments are 
5 subject to relapse. Therefore, notwithstanding what has been reported relating to the 
use of anti-CD20 antibodies and anti-CD22 antibodies in therapeutic regimens, it 
would be an advantage if novel therapeutic regimens could be developed, especially 
combination therapies that provide for enhanced therapeutic efficacy. In particular, it 
would be advantageous if novel therapies could be developed that prevent or reduce 
10 disease relapse in patients treated with RTTUXAN® or other anti-CD20 antibody 
therapeutic regimens. 

Preferred Embodiments of the Invention 

It is an embodiment of the invention to provide a novel therapeutic regimen 
1 5 that comprises the administration of an anti-CD20 monoclonal antibody or fragment 
thereof, and the administration of a radiolabeled anti-CD22 monoclonal antibody or 
fragment. 

It is another embodiment of the invention to provide a novel therapeutic 
regimen involving the initial administration of RTTUXAN®, followed by the 
20 administration of a radiolabeled anti-CD22 monoclonal antibody, or fragment thereof. 
It is another embodiment of the invention to provide a novel therapeutic 
regimen for the treatment of B-cell malignancies and cancers, especially B-cell 
leukemias or lymphomas, comprising the administration of RITUXAN®, followed by 
a radiolabeled humanized anti-CD22 antibody. 
25 It is another embodiment of the invention to provide novel methods for the 

treatment of autoimmune diseases and transplant comprising the administration of 
RITUXAN®, followed by a radiolabeled anti-CD22 antibody. 

It is another embodiment of the invention to provide novel methods of 
inhibiting B-cell immune responses, especially in protein, gene or cell therapy, or in 
30 the treatment of allergic disorders by the combined ao!mimstration of an anti-CD20 
antibody and a radiolabeled anti-CD22 antibody. 



6 



WO 01/97858 



PCT/US01/18939 



Summary of the Invention 

The present invention relates to methods of treating patients having diseases 
wherein inhibiting and/or depleting and/or killing and/or blocking the formation of B- 
cells is therapeutically desirable, especially B-cell malignancies and leukemias, as 
5 well as autoimmune diseases, transplant, allergic disorders, inflammatory disorders 
and gene or cell therapy, and other conditions wherein B-cell immunity is desirably 
suppressed, ha a preferred embodiment, the present invention relates to the treatment 
of B-cell lymphomas and leukemias, especially non-Hodgkin's lymphoma (NHL). 
Essentially, the subject therapeutic regimen will comprise the admimstration 
10 of a cold anti-CD20 antibody or fragment, and a hot (radiolabeled) anti-CD22 
antibody or fragment. The anti-CD20 antibody and the radiolabeled anti-CD22 
antibody can be administered in combination or separately, and in either order. 
Preferably, the anti-CD20 antibody will be administered first, in sufficient amounts to 
effect B-cell depletion, followed by the administration of a radiolabeled anti-CD22 
15 antibpdy. 

Preferably, this combination will affect synergistic results vis-a-vis the use of 
the cold anti-CD20 or radiolabeled anti-CD22 antibody or fragment alone. In a 
particularly preferred embodiment, this combination will provide for enhanced killing 
or depletion of tumorigenic B cells because the cold anti-CD20 antibody initially 
20 clears most CD20 expressing cells and the radiolabeled anti-CD22 antibody clears 
substantially all remaining tumorigenic B cells. Optionally, the combination therapy 
may further include the use of a radiolabeled anti-CD20 antibody, e.g. radiolabeled 
2B8 (Zevalin®). 

In another preferred embodiment, this combination will prevent or inhibit 
25 relapse in patients with B cell malignancies, e.g. non-Hodgkin's lymphoma, vis-a-vis 
current Rituxan®-based therapeutic regimens. 

Detailed Description of the Invention 

In order to clearly describe the invention, the following definitions are 
30 provided. 



7 



WO 01/97858 PCT/US01/18939 
Definitions 

Units, prefixes, and symbols can be denoted in their Si accepted form. 
Numeric ranges are inclusive of the numbers defining the range. Unless otherwise 
indicated, nucleic acids are written left to right in 5' to 3' orientation; amino acid 
5 sequences are written left to right in amino to carboxy orientation. The headings 
provided herein are not limitations of the various aspects or embodiments of the 
invention which can be had by reference to the specification as a whole. Accordingly, 
the terms defined immediately below are more fully defined by reference to the 
specification in its entirety. 

10 The term "antibody" as used herein is intended to include immunoglobulins 

and fragments thereof which are specifically reactive to the designated protein or 
peptide thereof. An antibody can include human antibodies, primatized antibodies, 
chimeric antibodies, bispecific antibodies, humanized antibodies, antibodies fused to 
other proteins or radiolabels, and antibody fragments. 

15 The term "antibody" herein is used in the broadest sense and specifically 

covers intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies 
(e.g. bispecific antibodies) formed from at least two intact antibodies, and antibody 
fragments so long as they exhibit the desired biological activity. 

"Antibody fragments" comprise a portion of an intact antibody, preferably 

20 comprising the antigen-binding or variable region thereof. Examples of antibody 

fragments include Fab, Fab', F(ab')2, and Fv fragments; diabodies; linear antibodies; 
single-chain antibody molecules; and multispecific antibodies formed from antibody 
fragments. Antibody fragments may be isolated using conventional techniques. For 
example, F(ab : )2 fragments can be generated by treating antibodies with pepsin. The 

25 resulting F(ab ! )2 fragment can be treated to reduce disulfide bridges to produce Fab 1 
fragments. 

"Native antibodies" are usually heterotetrameric glycoproteins of about 
150,000 daltons, composed of two identical light (L) chains and two identical heavy 
(H) chains. Each light chain is linked to a heavy chain by one covalent disulfide 
30 bond, while the number of disulfide linkages varies among the heavy chains of 
different immunoglobulin isotypes. Each heavy and light chain also has regularly 
spaced intrachain disulfide bridges. Each heavy chain has at one end a variable 



8 



WO 01/97858 



PCT/US01/18939 



domain (VH) followed by a number of constant domains. Each light chain has a 
variable domain at one end (VL) and a constant domain at its other end; the constant 
domain of the light chain is aligned with the first constant domain of the heavy chain, 
and the light-chain variable domain is aligned with the variable domain of the heavy 
5 chain. Particular amino acid residues are believed to form an interface between the 
light chain and heavy chain variable domains. 

The term "variable" refers to the fact that certain portions of the variable 
domains differ extensively in sequence among antibodies and are used in the binding 
and specificity of each particular antibody for its particular antigen. However, the 

10 variability is not evenly distributed throughout the variable domains of antibodies. It 
is concentrated in three segments called hypervariable regions both in the light chain 
and the heavy chain variable domains. The more highly conserved portions of variable 
domains are called the framework regions (FRs). The variable domains of native 
heavy and light chains each comprise four FRs, largely adopting a 13-sheet 

1 5 configuration, connected by three hypervariable regions, which form loops 
connecting, and in some cases forming part of, the B -sheet structure. The 
hypervariable regions in each chain are held together in close proximity by the FRs 
and, with the hypervariable regions from the other chain, contribute to the formation 
of the antigen-binding site of antibodies (see Kabat et al, Sequences of Proteins of 

20 Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, 
Bethesda, MD. (1 99 1)). The constant domains are not involved directly in binding an 
antibody to an antigen, but exhibit various effector functions, such as participation of 
the antibody in antibody dependent cellular cytotoxicity (ADCC). 

Papain digestion of antibodies produces two identical antigen-binding 

25 fragments, called "Fab" fragments, each with a single antigen-binding site, and a 
residual "Fc" fragment, whose name reflects its ability to crystallize readily. Pepsin 
treatment yields an F(ab')2 fragment that has two antigen-binding sites and is still 
capable of cross-linking antigen. 

"Fv" is the minimum antibody fragment which contains a complete antigen- 

30 recognition and antigen-binding site. This region consists of a dimer of one heavy 
chain and one light chain variable domain in tight, non-covalent association. It is in 
this configuration that the three hypervariable regions of each variable domain interact 



9 



WO 01/97858 PCT/US01/18939 

to define an antigen-binding site on the surface of the VH-VL dimer. Collectively, the 
six hypervariable regions confer antigen-binding specificity to the antibody. However, 
even a single variable domain (or half of an Fv comprising only three hypervariable 
regions specific for an antigen) has the ability to recognize and bind antigen, although 
5 at a lower affinity than the entire binding site. 

The Fab fragment also contains the constant domain of the light chain and the 
first constant domain (CHI) of the heavy chain. Fab' fragments differ from Fab 
fragments by the addition of a few residues at the carboxy terminus of the heavy chain 
CHI domain including one or more cysteines from the antibody hinge region. Fab'-SH 

10 is the designation herein for Fab' in which the cysteine residue(s) of the constant 

domains bear at least one free thiol group. F(ab')Z antibody fragments originally were 
produced as pairs of Fab' fragments which have hinge cysteines between them. Other 
chemical couplings of antibody fragments are also known. 

The "light chains" of antibodies (immunoglobulins) from any vertebrate 

15 species can be assigned to one of two clearly distinct types, called kappa and lambda, 
based on the amino acid sequences of their constant domains. 

Depending on the amino acid sequence of the constant domain of their heavy 
chains, antibodies can be assigned to different classes. There are five major classes of 
intact antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further 

20 divided into subclasses (isotypes), e.g., IgGI, IgG2, IgG3, IgG4, IgA, and IgA2. The 
heavy-chain constant domains that correspond to the different classes of antibodies 
are called alpha, delta, epsilon, gamma and mu, respectively. Preferably, the 
heavy-chain constant domains will complete the gamma- 1, gamma-2, gamma-3 and 
gamma-4 constant region. Preferably, these constant domains will also comprise 

25 modifications to enhance antibody stability such as the P and E modification disclosed 
in U.S. Patent No. 6,01 1,138 incorporated by reference in its entirety herein. The 
subunit structures and three dimensional configurations of different classes of 
immunoglobulins are well known. 

"Single-chain Fv" or "scFv" antibody fragments comprise the VH and VL 

30 domains of antibody, wherein these domains are present in a single polypeptide chain. 
Preferably, the Fv polypeptide further comprises a polypeptide linker between the VH 
and VL domains which enables the scFv to form the desired structure for antigen 



10 



WO 01/97858 



PCT/US01/18939 



binding. For a review of scFv see Pluckthun in The Pharmacology of Monoclonal 
Antibodies, vol. 113, Rosenburg and Moore eds., Springer-Verlag, New York, pp. 
269-315 (1994). 

The term "diabodies" refers to small antibody fragments with two 
5 antigen-binding sites, which fragments comprise a heavy-chain variable domain (VH) 
connected to a light chain variable domain (VL) in the same polypeptide chain (VH - 
VL). By using a linker that is too short to allow pairing between the two domains on 
the same chain, the domains are forced to pair with the complementary domains of 
another chain and create two antigen-binding sites. Diabodies are described more fully 

10 in, for example, EP 404,097; WO 93/1 1161; and Hollinger et al, Proc. Natl. Acad. 
Sci. USA, 90:6444-6448 (1993). 

The term "monoclonal antibody" as used herein refers to an antibody obtained 
from a population of substantially homogeneous antibodies, i.e., the individual 
antibodies comprising the population are identical except for possible naturally 

15 occurring mutations that may be present in rninor amounts. Monoclonal antibodies are 
highly specific, being directed against a single antigenic site. Furthermore, in contrast 
to conventional (polyclonal) antibody preparations which typically include different 
antibodies directed against different determinants (epitopes), each monoclonal 
antibody is directed against a single determinant on the antigen, hi addition to their 

20 specificity, the monoclonal antibodies are advantageous in that they are synthesized 
by the hybridoma culture, uncxmtarninated by other immunoglobulins. The modifier 
"monoclonal" indicates the character of the antibody as being obtained from a 
substantially homogeneous population of antibodies, and is not to be construed as 
requiring production of the antibody by any particular method. For example, the 

25 monoclonal antibodies to be used in accordance with the present invention may be 
made by the hybridoma method first described by Kohler et al, Nature, 256:495 
(1975), or may be made by recombinant DNA methods (see, e.g.,\J.S. Patent No. 
4,816,567). The "monoclonal antibodies" may also be isolated from phage antibody 
libraries using the techniques described in Clackson et al, Nature, 3 52:624-628 

30 (1991) and Marks et al, J. Mol Biol, 222:581-597 (1991), for example. 

By "humanized antibody" is meant an antibody derived from a non-human 
antibody, typically a murine antibody, that retains or substantially retains the antigen- 



11 



WO 01/97858 



PCT/US01/18939 



binding properties of the parent antibody, but which is less immunogenic in humans. 
This may be achieved by various methods, including (a) grafting the entire non-human 
variable domains onto human constant regions to generate chimeric antibodies; (b) 
grafting only the non-human complementarity determining regions (CDRs) into 
5 human framework and constant regions with or without retention of critical 

framework residues; and (c) transplanting the entire non-human variable domains, but 
"cloaking" them with a human-like section by replacement of surface residues. Such 
methods are disclosed in Morrison et al., Proc. Natl. Acad. Sci. 81 : 685 1-5 (1984); 
Morrison et al, Adv. Immunol. 44: 65-92 (1988); Verhoeyen et al, Science 239: 

10 1534-1536 (1988); Padlan, Molec. Immun. 28: 489-498 (1991); and Padlan, Molec. 
Immun. 31: 169-217 (1994), all of which are hereby incorporated by reference in their 
entirety. Humanized anti-CD40L antibodies can be prepared as described in U.S. 
Patent Application No. 08/554,840 filed November 7, 1995 also incorporated herein 
by reference in its entirety. 

15 By "human antibody" is meant an antibody containing entirely human light 

and heavy chain as well as constant regions, produced by any of the known standard 
methods. 

By "primatized antibody" is meant a recombinant antibody which has been 
engineered to contain the variable heavy and light domains of a monkey (or other 

20 primate) antibody, in particular, a cynomolgus monkey antibody, and which contains 
human constant domain sequences, preferably the human immunoglobulin gamma 1 
or gamma 4 constant domain (or PE variant). The preparation of such antibodies is 
described in Newman et al, Biotechnology, 10: 1458-1460 (1992); also in commonly 
assigned 08/379,072, 08/487,550, or 08/746,361, all of which are incorporated by 

25 reference in their entirety herein. These antibodies have been reported to exhibit a 
high degree of homology to human antibodies, i.e., 85-98%, display human effector 
functions, have reduced immunogenicity, and may exhibit high affinity to human 
antigens. 

By "antibody fragment" is meant an fragment of an antibody such as Fab, 
30 F(ab')2, Fab' and scFv. 

By "chimeric antibody" is meant an antibody containing sequences derived 
from two different antibodies, which typically are of different species. Most typically, 



12 



WO 01/97858 



PCT/US01/18939 



chimeric antibodies comprise human and murine antibody fragments, and generally 
human constant and murine variable regions. 

"B Cell Depleting Antibody" therein is an antibody or fragment that upon 
administration, results in demonstrable B cell depletion. Typically, such antibody will 
5 bind to a B cell antigen or B cell marker expressed on the surface of a B cell. 

Preferably, such antibody, after adrniriistration, typically within about several days or 
less, will result in a depletion of B cell number by about 50% or more. In a preferred 
embodiment, the B cell depleting antibody will be RITUXAN® (a chimeric anti- 
CD20 antibody) or one having substantially the same or at least 20-50% the cell 
1 0 depleting activity of RITUXAN®, over the same time period, preferably at least 90% 
thereof. 

A "B cell surface marker" or "B cell target" or "B cell antigen" is an antigen 
expressed on the surface of a B cell which can be targeted with an antagonist which 
binds thereto. Exemplary B cell surface markers include the CD10, CD19, CD20, 

15 CD21, CD22, CD23, CD24, CD37, CD53, CD72, CD73, CD74, CDw75, CDw76, 
CD77, CDw78, CD79a, CD79b, CD80, CD81, CD82, CD83, CDw84, CD85 and 
CD86 leukocyte surface markers. A preferred B cell surface marker is preferentially 
expressed on B cells compared to other non-B cell tissues of a mammal and may be 
expressed on both precursor B cells and mature B cells. 

20 The "CD20" antigen is a -35 kDa, non-glycosylated phosphoprotein found on 

the surface of greater than 90% of B cells from peripheral blood or lymphoid organs. 
CD20 is expressed during early pre-B cell development and remains until plasma cell 
differentiation. CD20 is present on both normal B cells as well as malignant B cells. 
Other names for CD20 in the literature include "B-lymphocyte-restricted antigen" and 

25 "Bp35". The CD20 antigen is described in Clark et al. PNAS (USA) 82:1766(1985). 

The "CD22" antigen refers to an antigen expressed on B cells, also known as 
"BL-CAM" and "LybB" that is involved in B cell signaling and an adhesion. (See 
Nitschke et al, Curr. Biol. 7:133 (1997); Stamenkovic et al, Nature 345:74 (1990)). 
This antigen is a membrane immunoglobulin-associated antigen that is tyrosine 

3 0 phosphorylated when membrane Ig is ligated. (Engel et al, J. Etyp. Med. 1 8 1 (4) : 1 52 1 
1586 (1995)). The gene encoding this antigen has been cloned, and its lg domains 
characterized. 



13 



WO 01/97858 



PCT/US01/18939 



A B cell "antagonist" is a molecule which, upon binding to a B cell surface 
marker, destroys or depletes B cells in a mammal and/or interferes with one or 
more B cell functions, e.g. by reducing or preventing a humoral response elicited by 
the B cell. The antagonist preferably is able to deplete B cells (i.e. reduce circulating 
5 B cell levels) in a mammal treated therewith. Such depletion may be achieved via 
various mechanisms such antibody-dependent cell-mediated cytotoxicity (ADCC) 
and/or complement dependent cytotoxicity (CDC), inhibition of B cell proliferation 
and/or induction of B cell death (e.g. via apoptosis). Antagonists included within the 
scope of the present invention include antibodies, synthetic or native sequence 
10 peptides and small molecule antagonists which bind to the B cell marker, optionally 
conjugated with or fused to a cytotoxic agent. 

"B cell depleting antibody" is an antibody that, upon in vivo adrninistration, 
reduces the number of circulating B cells. Preferably, depletion will occur within 
about 24 hours of administration to levels which are at least 50% depletion or more. 
15 Most preferably, a cold anti-CD20 antibody will deplete B cells substantially as 

efficiently (within about 80-90% of the level of B cell depletion within same time) as 
Rituxan®. 

"Antibody-dependent cell-mediated cytotoxicity" and "ADCC" refer to a cell 
mediated reaction in which nonspecific cytotoxic cells that express Fc receptors 

20 (FcRs) (e.g. Natural Killer (NK) cells, neutrophils, and macrophages) recognize 
bound antibody on a target cell and subsequently cause lysis of the target cell. The 
primary cells for mediating ADCC, NK cells, express FcyRTH only, whereas 
monocytes express FcyRI, FcyRH and FcyRHI. FcR expression on hematopoietic cells 
in summarized is Table 3 on page 464 of Ravetch and Kinet, Annu. Rev. Immunol 

25 9:457-92 (1 99 1 ). To assess ADCC activity of a molecule of interest, an in vitro 

ADCC assay, such as that described in US Patent No. 5,500,362 or 5,821,337 may be 
performed. Useful effector cells for such assays include peripheral blood mononuclear 
cells (PBMC) and Natural Killer (NK) cells. Alternatively, or additionally, ADCC 
activity of the molecule of interest maybe assessed in vivo, e.g., in a animal model 

30 such as that disclosed in Clynes et al. PNAS (USA) 95 :652-656 (1 998). 

"Human effector cells" are leukocytes which express one or more FcRs and 
perform effector functions. Preferably, the cells express at least FcyRTJI and perform 



14 



WO 01/97858 



PCTYUS01/18939 



ADCC effector function. Examples of human leukocytes which mediate ADCC 
include peripheral blood mononuclear cells (PBMC), natural killer (NK) cells, 
monocytes, cytotoxic T cells and neutrophils; with PBMCs and NK cells being 
preferred. The effector cells may be isolated from a native source thereof, e.g. from 
5 blood or PBMCs as described herein. 

The terms "Fc receptor" or "FcR" are used to describe a receptor that binds to 
the Fc region of an antibody. The preferred FcR is a native sequence human FcR. 
Moreover, a preferred FcR is one which binds an IgG antibody (a gamma receptor) 
and includes receptors of the FcyRI, FcyRII, and FcyRII subclasses, including allelic 

1 0 variants and alternatively spliced forms of these receptors. FcyRII receptors include 
FcyRUA (an "activating receptor") and FcyRUB (an "inhibiting receptor"), which 
have similar amino acid sequences that differ primarily in the cytoplasmic domains 
thereof. Activating receptor FcyRCA contains an immunoreceptor tyrosine-based 
activation motif (ITAM) in its cytoplasmic domain. Inhibiting receptor FcyRUB 

15 contains an immunoreceptor tyrosine-based inhibition motif (ITIM) in its cytoplasmic 
domain, (see review M. in Daeon, Annu. Rev. Immunol. 15:203-234 (1997)). FcRs 
are reviewed in Ravetch and Kinet, Annu. Rev. Immunol. 9:457-92 (1991); Capel et 
al., hnmunomethods 4:25-34 (1994); and de Haas et al., J. Lab. Clin. Med. 
126:330-41 (1995). Other FcRs, including those to be identified in the future, are 

20 encompassed by the term "FcR" herein. The term also includes the neonatal receptor, 
FcRn, which is responsible for the transfer of maternal IgGs to the fetus (Guyer et al, 
J. Immunol. 117:587 (1976) and Kim et al., J. Immunol. 24:249 (1994)). 

"Complement dependent cytotoxicity" or "CDC" refers to the ability of a 
molecule to lyse a target in the presence of complement. The complement activation 

25 pathway is initiated by the binding of the first component of the complement system 
(Clq) to a molecule (e.g. an antibody) complexed with a cognate antigen. To assess 
complement activation, a CDC assay, e.g. as described in Gazzano-Santoro et al., J. 
Immunol. Methods 202:163 (1996), maybe performed. 

"Growth inhibitory" antagonists are those which prevent or reduce 

30 proliferation of a cell expressing an antigen to which the antagonist binds. For 

example, the antagonist may prevent or reduce proliferation of B cells in vitro and/or 
in vivo. 



15 



WO 01/97858 



PCT/US01/18939 



Antagonists which "induce apoptosis" are those which induce programmed 
cell death, e.g. of a B cell, as determined by binding of annexin V, fragmentation of 
DNA, cell shrinkage, dilation of endoplasmic reticulum, cell fragmentation, and/or 
formation of membrane vesicles (called apoptotic bodies). 
5 The term "hypervariable region" when used herein refers to the amino acid 

residues of an antibody which are responsible for antigen-binding. The hypervariable 
region comprises amino acid residues from a "complementarity determining region" 
or "CDR" (e.g. residues 24-34 (LI), 50-56 (L2) and 89-97 (L3) in the light chain 
variable domain and 31-35 (HI), 50-65 (H2) and 95-102 (H3) in the heavy chain 

1 0 variable domain; Kabat et al, Sequences of Proteins of Immunological Interest, 5th 
Ed. Public Health Service, National Institutes of Health, Bethesda, MD. (1991)) 
and/or those residues from a "hypervariable loop" (e.g. residues 26-32 (LI), 50-52 
(L2) and 91-96 (L3) in the light chain variable domain and 26-32 (HI), 53-55 (H2) 
and 96-101 (H3) in the heavy chain variable domain; Chothia and Lesk.l. Mol. Biol. 

15 196:901-917 (1987)). "Framework" or 'TR" residues are those variable domain 
residues other than the hypervariable region residues as herein denned. 

An antagonist "which binds" an antigen of interest, e.g. a B cell surface 
marker, is one capable of binding that antigen with sufficient affinity such that the 
antagonist is useful as a therapeutic agent for targeting a cell, i.e. a B cell, expressing 

20 the antigen. 

An "anti-CD20 antibody" herein is an antibody that specifically binds CD20 
antigen, preferably human CD20, having measurable B cell depleting activity, 
preferably having at least about 10%, more preferably at least 50%, and still more 
preferably at least 90%, the B cell depleting activity of RITUXAN® (see U.S. Patent 

25 No. 5,736,137, incorporated by reference herein in its entirety). 

An "anti-CD22 antibody" herein is an antibody that specifically binds CD22 
antigen, preferably human CD22, having measurable B cell depleting activity, 
preferably having at least about 10% the B cell depleting activity of RITUXAN® (see 
U.S. Patent No. 5,736,137, incorporated by reference herein in its entirety). 

30 Specific examples of antibodies which bind the CD20 antigen include': 

"Rituximab" ("RITUXAN®") (US PatentNo. 5,736,137, expressly incorporated 
herein by reference); yttrium-[90]-labeled 2B8 murine antibody "Y2B8" (US Patent 



16 



WO 01/97858 PCT/US01/18939 

No. 5,736,137, expressly incorporated herein by reference); murine IgG2a"Bl" 
optionally labeled with 131 I labeled Bl antibody (BEXXARTM) (US Patent No. 
5,595,721, expressly incorporated herein by reference); murine monoclonal antibody 
"1F5" (Press et al. Blood 69(2):584-591 (1987); and "chimeric 2H7" antibody (US 
5 Patent No. 5,677, 1 80, expressly incorporated herein by reference). 

Specific examples of antibodies which bind CD22 include Lymphocide™ 
reported by Immunomedics, now in clinical trials for non-Hodgkin's lymphoma. 

The terms "rituximab" or "PJTUXAN®" herein refer to the genetically 
engineered chimeric murme/human monoclonal antibody directed against the CD20 
10 antigen and designated "C2B8" in US Patent No. 5,736,B7, expressly incorporated 
herein by reference. The antibody is an IgGI kappa immunoglobulin amtaining 
murine light and heavy chain variable region sequences and human constant region 
sequences. Rituximab has a binding affinity for the CD20 antigen of approximately 
8.0 nM. 

15 An "isolated" antagonist is one which has been identified and separated and/or 

recovered from a component of its natural environment. Contaminant components of 
its natural environment are materials which would interfere with diagnostic or 
therapeutic uses for the antagonist, and may include enzymes, hormones, and other 
proteinaceous or nonproteinaceous solutes. In preferred embodiments, the antagonist 

20 will be purified (1) to greater than 95% by eight of antagonist as determined by the 
Lowry method, and most preferably more than 99% by weight, (2) to a degree 
sufficient to obtain at least 15 residues of N-terminal or internal arnino acid sequence 
by use of a spinning cup sequenator, or (3) to homogeneity by SDS-PAGE under 
reducing or nonreducing conditions using Coomassie blue or, preferably, silver stain. 

25 Isolated antagonist includes the antagonist in situ within recombinant cells since at 
least one component of the antagonist's natural environment will not be present. 
Ordinarily, however, isolated antagonist will be prepared by at least one purification 
step. 

"Mammal" for purposes of treatment refers to any animal classified as a 
30 mammal, including humans, domestic and farm animals, and zoo, sports, or pet 
animals, such as dogs, horses, cats, cows, etc. Preferably, the mammal is human. 
"Treatment" refers to both therapeutic treatment and prophylactic or 



17 



WO 01/97858 



PCT/US01/18939 



preventative measures. Those in need of treatment include those already with the 
disease or disorder as well as those in which the disease or disorder is to be prevented. 
Hence, the mammal may have been diagnosed as having the disease or disorder or 
maybe predisposed or susceptible to the disease. 
5 B Cell Malignancy 

According to the present invention this includes any B cell malignancy, e.g., B 
cell lymphomas and leukemias. Preferred examples include Hodgkin's disease (all 
forms, e.g., relapsed Hodgkin's disease, resistant Hodgkin's disease) non-Hodgkin's 
lymphomas (low grade, intermediate grade, high grade, and other types). Examples 

10 include small lymphocytic/B cell chronic lymphocytic leukemia (SLL/B-CLL), 
lymhoplasmacytoid lymphoma (LPL), mantle cell lymphoma (MCL), follicular 
lymphoma (FL), diffuse large cell lymphoma (DLCL), Burkitt's lymphoma (BL), 
AIDS- related lymphomas, monocytic B cell lymphoma, angioimmunoblastic 
lymphoadenopathy, small lymphocytic, follicular, diffuse large cell, diffuse small 

15 cleaved cell, large cell immunoblastic lymphoblastoma, small, non-cleaved, Burkitt's 
and non-Burkitt's, follicular, predominantly large cell; follicular, predominantly small 
cleaved cell; and follicular, mixed small cleaved and large cell lymphomas. See, 
Gaidono et al., "Lymphomas", IN CANCER: PRINCIPLES & PRACTICE OF 
1 ONCOLOGY, Vol. 2: 213 1-2145 (DeVita et al., eds., 5 th ed. 1997). 

20 Other types of lymphoma classifications include immunocytomal 

Waldenstrom's MALT-type/monocytoid B cell, mantle cell lymphoma B-CLL/SLL, 
diffuse large B-cell lymphoma, follicular lymphoma, and precursor B-LBL. 

As noted, B cell malignancies further include especially leukemias such as 
ALI^L3 (Burkitt's type leukemia), chronic lymphocytic leukemia (CLL), chronic 

25 leukocytic leukemia, acute myelogenous leukemia, acute lymphoblastic leukemia, 
chronic lymphocytic leukemia, chronic myelogenous leukemia, lymphoblastic 
leukemia, lymphocytic leukemia, monocytic leukemia, myelogenous leukemia, and 
promyelocytic leukemia and monocytic cell leukemias. 

"Autoimmune disease" herein includes any autoimmune disease wherein 

30 elimination or depletion or inhibition of the activity or proliferation of B cells is 

therapeutically beneficial. Such autoimmune diseases will include in particular T and 
B cell mediated autoimmune diseases. Examples thereof include: the treatment or 



18 



WO 01/97858 



PCTVUS01/18939 



prevention of autoimmune, inflammatory, proliferative and hyperproliferative 
diseases, and of cutaneous manifestations of immunologically medicated diseases 
(e.g., rheumatoid arthritis, lupus erythematosus, systemic lupus erythematosus, 
Hashimotos thyroiditis, multiple sclerosis, myasthenia gravis, type 1 diabetes, uveitis, 
5 nephrotic syndrome, psoriasis, atopical dermatitis, contact dermatitis and further 
eczematous dermatitides, seborrheic dermatitis, Lichen planus, Pemphigus, bullous 
pemphigus, Epidermolysis bullosa, urticaria, angioedemas, vasculitides, erythema, 
cutaneous eosinophilias, Alopecia areata, etc.); the treatment of reversible obstructive 
airways disease, intestinal inflammations and allergies (e.g., inflammatory bile 

10 disease, Coeliac disease, proctitis, eosinophilia gastroenteritis, mastocytosis, Crohn's 
disease and ulcerative colitis), food-related allergies (e.g., migraine, rhinitis and 
eczema), and other types of allergies. 

Also, the subject combination therapy is useful for treating malignancies, 
particularly solid tumors or late stage malignancies wherein B cells promote tumor 

1 5 growth, maintenance and/or metastasis but wherein B cells are not themselves the 
origin of the malignancy (not B cell malignancy such as non-Hodgkin's lymphoma). 

Cell therapy includes any therapy wherein a potentially immunogenic cell is 
introduced into a subject, e.g. isogeneic, allogeneic or xenogeneic, which potentially 
may contain a tetrologus gene, e.g. one encoding a therapeutic polypeptide. 

20 Gene therapy includes any therapy wherein a DNA or RNA sequence is 

introduced that modulates (inhibits or enhances) or provides for the expression of a 
gene normally or not normallyl expressed, e.g. one involved in a disease. Typically, 
the DNA or RNA will be comprised in a vector, e.g. plasmid, virus or in the genome 
of a cell, e.g. mammalian cell. Alternatively, the DNA or RNA may be "naked" or 

25 comprised in a stabilizing or targeting material, e.g. liposome. Examples include 
adenoviral, poxviruses, and other viral vectors, liposomal DNA formulations, etc. 

The expression "therapeutically effective amount" refers to an amount of the 
naked antibody or radiolabeled antibody which is effective for preventing, 
ameliorating or treating the disease in question, e.g. B cell malignancy. 

30 The term "immunosuppressive agent" as used herein for adjunct therapy refers 

to substances that act to suppress or mask the immune system of the mammal being 
treated herein. This would include substances that suppress cytokine production, 



19 



WO 01/97858 



PCT7US01/18939 



downregulate or suppress self-antigen expression, or mask the MHC antigens. 
Examples of such agents include 2-ammo-6-aryl-5-substimtedpyrirrudines (see U.S. 
Pat. No. 4,665,077, the disclosure of which is incorporated herein by reference), 
azatbioprine; cyclophosphamide; bromocryptine; danazol; dapsone; glutaraldehyde 
5 (which masks the MHC antigens, as described in U.S. Pat. No. 4, 120,649); 

anti-idiotypic antibodies for MHC antigens and MHC fragments; cyclosporin A; 
steroids such as glucocorticosteroids, e.g., prednisone, methylprednisolone, and 
dexamethasone; cytokine or cytokine receptor antagonists including anti-interferon-a, 
P- or 8-antibodies, anti-tumor necrosis factor-a antibodies, anti-tumor necrosis 

10 factor-P antibodies, anti-interleukin-2 antibodies and anti-IL-2 receptor antibodies; 
anti-LFA-1 antibodies, including anti-CDl la and anti-CD 18 antibodies; anti-L3T4 
antibodies; heterologous anti-lymphocyte globulin; pan-T antibodies, preferably 
anti-CD3 or anti-CD4/CD4a antibodies; soluble peptide containing a LFA-3 binding 
domain (WO 90/08187 published 7/26/90), streptolanase; TGF-P; streptodornase; 

15 RNA or DNA from the host; FK506; RS-61443; deoxyspergualin; rapamycin; T-cell 
receptor (Cohen et al, U.S. Pat. No. 5,1 14,721); T-cell receptor fragments (Offher et 
al, Science, 251: 430-432 (1991); WO 90/11294; Laneway, Nature, 341: 482 (1989); 
and WO 91/01133); and T cell receptor antibodies (EP 340,109) such as T10B9. 

The term "cytotoxic agent" as used herein refers to a substance that inhibits or 

20 prevents the function of cells and/or causes destruction of cells. The term is intended 
to include radioactive isotopes (e.g. 123 1, 125 1, 131 I, lu Tn, 131 Tn, 52 P, "C, 67 Cu, 2u At, 
,77 Lu, "Y, ,86 Re, 212 Pb, 212 Bi, 47 Sc, 105 Rh, 104 Pd, 153 Sm, 188 Re, 199 Au, 2n At and 213 Bi), 
chemotherapeutic agents, and toxins such as small molecule toxins or enzymatically 
active toxins of bacterial, fungal, plant or animal origin, or fragments thereof. 

25 A "chemotherapeutic agent" is a chemical compound useful in the treatment of 

cancer. Examples of chemotherapeutic agents include alkylating agents such as 
thiotepa and cyclosphosphamide (CYTOXAN™); alkyl sulfonates such as busulfan, 
improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, 
and uredopa; ethylenimines and memylamelamines mcluding dfretarnine, 

30 triemylenemelamine, trietylenephosphoramide, rriethylenethiophosphaorarnide and 
trimethylolomelarnime nitrogen mustards such as chlorambucil, chlomaphazine, 
cholophosphamide, estramustine, ifosfamide, mecUoremamine, mecWoremamine 



20 



WO 01/97858 PCT/US01/18939 

oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, 
trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, 
fotemustine, lomustine, nimustine, ranimustine; antibiotics such as aclacinomysins, 
actinomycin, authramycin, azaserine, bleomycins, cactinomycin, calicheamicin, 
5 carabicin, carminomycin, carzinophilin, chromomycins, dactmomycin, daunorubicin, 
detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin, epirubicin, esorubicin, 
idarubicin, marcellomycin, mitomycins, mycophenolic acid, nogalamycin, 
olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, 
streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; 

10 anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid 

analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs 
such as fludarabine, 6-mercaptopurine, thiamiprine, unoguanine; pyrirmdme analogs 
such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyiuidine, 
doxifluridine, enocitabine, floxuridine, 5-FU; androgens such as calusterone, 

15 dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals 
such as ammoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic 
acid; aceglatone; aldophosphamide glycoside; arninolevulinic acid; amsacrine; 
bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; 
elfoiruthine; elliptinium acetate; etoglucid; gallium nitrate; hydroxyurea; Ientinan; 

20 lomdamine; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; 

phenamet; pirarubicin; podophyllinic acid'; 2-ethylhydrazide; procarbazine; PSK®; 
razoxane; sizofiran; spirogermanium; tenua?onic acid; triaziquone; 2, 
2^2"-tricUorotriemylamine; urethan; vindesine; dacarbazine; mannomustine; 
mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); 

25 cyclophosphamide; thiotepa; taxoids, e.g. paclitaxel (TAXOL®, Bristol-Myers Squibb 
Oncology, Princeton, NJ) and doxetaxel (Taxotere, Rhone-Poulenc Rorer, Antony, 
France); chlorambucil; gemcitabine; 6-tmoguanine; mercaptopurine; methotrexate; 
platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide 
(VP- 16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; 

30 novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; CPT1 1; 
topoisomerase inhibitor RFS 2000; d^fluoromemylormthine (DMFO); retinoic acid; 
esperamicins; capecitabine; and pharmaceutically acceptable salts, acids or derivatives 



21 



WO 01/97858 



PCT/US01/18939 



of any of the above. Also included in this definition are anti-hormonal agents that act 
to regulate or inhibit hormone action on tumors such as anti-estrogens including for 
example tamoxifen, raloxifene, aromatase inhibiting 4(5)-imidazoles, 
4-hydroxytamoxifen, trioxifene, keoxifene, LY1 17018, onapristone, and toremifene 
5 (Fareston); and antiandrogens such as flutamide, nilutamide, bicalutamide, leuprohde, 
and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the 
above. 

The term "cytokine" is a generic term for proteins released by one cell 
population which act on another cell as intercellular mediators. Examples of such 

10 cytokines are lymphokines, monokines, and traditional polypeptide hormones. 

Included among the cytokines are growth hormone such as human growth hormone, 
N-methionyl human growth hormone, and bovine growth hormone; parathyroid 
hormone; thyroxine; insulin; proinsulin; relaxin; prorelaxin; glycoprotein hormones 
such as follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH), and 

15 luteinizing hormone (LH); hepatic growth factor; fibroblast growth factor; prolactin; 
placental lactogen; tumor necrosis factor-oc and -p; mullerian-inhibiting substance; 
mouse gonadotropin-associated peptide; inhibin; activin; vascular endothelial growth 
factor; integrin; thrombopoietin (TPO); nerve growth factors such as NGF-13; 
platelet-growth factor; transforming growth factors (TGFs) such as TGF-a and 

20 TGF-P; insulin-like growth factor-I and -II; erythropoietin (EPO); osteoinductive 

factors; interferons such as interferon-a, -p, and -y; colony stimulating factors (CSFs) 
such as macrophage-CSF (M-CSF); granulocytemacrophage-CSF (GM-CSF); and 
granulocyte-CSF (G-CSF); interleukins (ILs) such as IL-1, IL-la, IL-2, IL-g, EL-4, 
IL-5, EL-6, lL-7, IL-8, IL-9, IL-1 1, IL-12, IL-15; a tumor necrosis factor such as 

25 TNF-ct or TNF-p; and other polypeptide factors including LIF and kit ligand (KL). As 
used herein, the term cytokine includes proteins from natural sources or from 
recombinant cell culture and biologically active equivalents of the native sequence 
cytokines. 

The term "prodrug" as used in this application refers to a precursor or 
30 derivative form of a pharmaceutically active substance that is less cytotoxic to tumor 
cells compared to the parent drug and is capable of being enzymatically activated or 
converted into the more active parent form. See, e.g., Wilman, "Prodrugs in Cancer 

22 



WO 01/97858 PCI7US01/18939 

Chemotherapy" Biochemical Society Transactions, 14, pp. 375-382, 615th Meeting 
Belfast (1986) and Stella et al, "Prodrugs: A Chemical Approach to Targeted Drug 
Delivery," Directed Drug Delivery, Borchardt et al, (ed.), pp. 247-267, Humana Press 
(1985). The prodrugs of this invention include, but are not limited to, 
5 phosphate-containing prodrugs, ttaophosphate-containing prodrugs, sulfate-containing 
prodrugs, pepn^e-containing prodrugs, D-amino acid-modified prodrugs, glycosylated 
prodrugs, 1 3-lactam-containing prodrugs, optionally substituted 
phenoxyacetamide-containing prodrugs or optionally substituted 
phenylacetamide-containing prodrugs, 5-fluorocytosine and other 5fluorouridine 

10 prodrugs which can be converted into the more active cytotoxic free drug. Examples 
of cytotoxic drugs that can be derivatized into a prodrug form for use in this invention 
include, but are not limited to, those chemotherapeutic agents described above. 

A "liposome" is a small vesicle composed of various types of lipids, 
phospholipids and/or surfactant which is useful for delivery of a drug (such as the 

15 antagonists disclosed herein and, optionally, a chemotherapeutic agent) to a mammal. 
The components of the liposome are commonly arranged in a bilayer formation, 
similar to the lipid arrangement of biological membranes. 

The term "package insert" is used to refer to instructions customarily included 
in commercial packages of therapeutic products, that contain information about the 

20 indications, usage, dosage, administration, contraindications and/or warnings 
concerning the use of such therapeutic products. 
Production of Antibodies 

The subject invention uses antibodies to CD20 and CD22. These antibodies 
will be provided by known methods. As noted, antibodies to both these antigens are 

25 well known. 

Exemplary techniques for the production of the antibodies used in accordance 
with the present invention are described. 
(i) Polyclonal antibodies 

Polyclonal antibodies are preferably raised in animals by multiple 
30 subcutaneous (sc) or intraperitoneal (ip) injections of the relevant antigen and an 
adjuvant. It may be useful to conjugate the relevant antigen to a protein that is 
immunogenic in the species to be immunized, e.g., keyhole limpet hemocyanin, serum 



23 



WO 01/97858 



PCT/US01/18939 



albumin, bovine thyroglobulin, or soybean trypsin inhibitor using a Afunctional or 
derivatizing agent, for example, maleirnidobenzoyl sulfosuccinimide ester 
(conjugation through cysteine residues), N-hydroxysuccinhnide (through lysine 
residues), glutaraldehyde, succinic anhydride, SOCl 2 , or R 1 N=C=NR, where R and R 1 
5 are different alkyl groups. 

Animals are immunized against the antigen, immunogenic conjugates, or 
derivatives by combining, e.g. 100 ug or 5 ug of the protein or conjugate (for rabbits 
or mice, respectively) with 3 volumes of Freund's complete adjuvant and injecting the 
solution intradermally at multiple sites. One month later the animals are boosted with 

10 1/5 to 1/10 the original amount of peptide or conjugate in Freund's complete adjuvant 
by subcutaneous injection at multiple sites. Seven to 14 days later the animals are bled 
and the serum is assayed for antibody titer. Animals are boosted until the titer 
plateaus. Preferably, the animal is boosted with the conjugate of the same antigen, but 
conjugated to a different protein and/or through a different cross-linking reagent. 

1 5 Conjugates also can be made in recombinant cell culture as protein fusions. Also, 
aggregating agents such as alum are suitably used to enhance the immune response. 
(ii) Monoclonal antibodies 

Monoclonal antibodies are obtained from a population of substantially 
homogeneous antibodies, i.e., the individual antibodies comprising the population are 

20 identical except for possible naturally occurring mutations that may be present in 
minor amounts. Thus, the modifier "monoclonal" indicates the character of the 
antibody as not being a mixture of discrete antibodies. 

For example, the monoclonal antibodies may be made using the hybridoma 
method first described by Kohler et al, Nature, 256:495 (1975), or may be made by 

25 recombinant DNA methods (U.S. Patent No. 4,816,567). 

In the hybridoma method, a mouse or other appropriate host animal, such as a 
hamster, is immunized as hereinabove described to elicit lymphocytes that produce or 
are capable of producing antibodies that will specifically bind to the protein used for 
immunization. Alternatively, lymphocytes may be immunized in vitro. Lymphocytes 

30 then are fused with myeloma cells using a suitable fusing agent, such as polyethylene 
glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and 
Practice, pp.59-1 03 (Academic Press, 1986)). 



24 



WO 01/97858 



PCT/US01/18939 



The hybridoma cells thus prepared are seeded and grown in a suitable culture 
medium that preferably contains one or more substances that inhibit the growth or 
survival of the unfused, parental myeloma cells. For example, if the parental myeloma 
cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or 
5 HPRT), the culture medium for the hybridomas typically will include hypoxanthine, 
arninopterin, and mymidine (HAT medium), which substances prevent the growth of 
HGPRT-deficient cells. 

Preferred myeloma cells are those that fuse efficiently, support stable 
high-level production of antibody by the selected antibody-producing cells, and are 

10 sensitive to a medium such as HAT medium. Among these, preferred myeloma cell 
lines are murine myeloma lines, such as those derived from MOPC-21 and MPC-1 1 
mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, 
CaUfornia USA, and SP-2 or X63-Ag8-653 cells available from the American Type 
Culture Collection, Manassas, Virginia, USA. Human myeloma and mouse-human 

15 heteromyeloma cell lines also have been described for the production of human 
monoclonal antibodies (Kozbor, J. Immunol., 133:300 1 (1984); Brodeur etal, 
Monoclonal Antibody Production Techniques and Applications, pp. 51-63 (Marcel 
Dekker, Inc., New York, 1987)). 

Culture medium in which hybridoma cells are growing is assayed for 

20 production of monoclonal antibodies directed against the antigen. Preferably, the 
binding specificity of monoclonal antibodies produced by hybridoma cells is 
determined by immunoprecipitation or by an in vitro binding assay, such as 
radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA). 
The binding affinity of the monoclonal antibody can, for example, be 

25 determined by the 30 Scatchard analysis of Munson et al, Anal Biochem., 107:220 
(1980). 

After hybridoma cells are identified that produce antibodies of the desired 
specificity, affinity, and/or activity, the clones maybe subcloned by limiting dilution 
procedures and grown by standard methods (Goding, Monoclonal Antibodies: 
30 Principles and Practice, pp.59-103 (Academic Press, 1986)). Suitable culture media 
for this purpose include, for example, D-MEM or RPML-1640 medium. In addition, 
the hybridoma cells may be grown in vivo as ascites tumors in an animal. 



25 



WO 01/97858 



PCT/US01/18939 



The monoclonal antibodies secreted by the subclones are suitably separated 
from the culture medium, ascites fluid, or serum by conventional immunoglobulin 
purification procedures such as, for example, protein A-Sepharose, hydroxylapatite 
chromatography, gel electrophoresis, dialysis, or affinity chromatography. 
5 DNA encoding the monoclonal antibodies is readily isolated and sequenced 

using conventional procedures (e.g., by using oligonucleotide probes that are capable 
of binding specifically to genes encoding the heavy and light chains of murine 
antibodies). The hybridoma cells serve as a preferred source of such DNA. Once 
isolated, the DNA may be placed into expression vectors, which are then transfected 

10 into host cells such as E. coli cells, simian COS cells, Chinese Hamster Ovary (CHO) 
cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to 
obtain the synthesis of monoclonal antibodies in the recombinant host cells. Review 
articles on recombinant expression in bacteria of DNA encoding the antibody include 
Skerra et al, Curr. Opinion in Immunol, 5:256-262 (1993) and Pluckthun, Immunol. 

15 Revs., 130:151-188 (1992). 

Another method of generating specific antibodies, or antibody fragments, 
reactive against a CD20 or CD22 is to screen expression libraries encoding 
immunoglobulin genes, or portions thereof, expressed in bacteria with a CD20 or 
CD22 protein or peptide. For example, complete Fab fragments, Vh regions and Fv 

20 regions can be expressed in bacteria using phage expression libraries. See for 

example, Ward et al, Nature 341: 544-546 (1989); Huse et al, Science 246: 1275- 
1281 (1989); and McCafferty et al, Nature 348: 552-554 (1990). Screening such 
libraries with, for example, a CD22 or CD20 peptide, can identify irnmunoglobulin 
fragments reactive with CD22 or CD20. Alternatively, the SCID-hu mouse (available 

25 from Genpharm) can be used to produce antibodies or fragments thereof. 

In a further embodiment, antibodies or antibody fragments can be isolated 
from antibody phage libraries generated using the techniques described in McCafferty 
et al, Nature, 348:552-554(1990). Clackson et al, Nature, 352:624-628 (1991) and 
Marks et al, J. Mol. Biol, 222:581-597 (1991) describe the isolation of murine and 

30 human antibodies, respectively, using phage libraries. Subsequent publications 
describe the production of high affinity (nM range) human antibodies by chain 
shuffling (Marks et al, Bio/Technology, 10:779-783 (1992)), as well as combinatorial 



26 



WO 01/97858 



PCT/US01/18939 



infection and in vivo recombination as a strategy for constructing very large phage 
libraries (Waterhouse et al, Nuc. Acids. Res., 21:2265-2266 (1993)). Thus, these 
techniques are viable alternatives to traditional monoclonal antibody hybridoma 
techniques for isolation of monoclonal antibodies. 
5 The DNA also may be modified, for example, by substituting the coding 

sequence for human heavy- and tight-chain constant domains in place of the 
homologous murine sequences (U.S. Patent No. 4,816,567; Morrison, et al, Proc. 
Natl Acad. ScL USA, 81:6851 (1984)), or by covalently joining to the immunoglobulin 
coding sequence all or part of the coding sequence for a non-immunoglobulin 

10 polypeptide. 

Typically, such non-immunoglobulin polypeptides are substituted for the 
constant domains of an antibody, or they are substituted for the variable domains of 
one antigencombining site of an antibody to create a chimeric bivalent antibody 
comprising one antigen-combining site having specificity for an antigen and another 

1 5 antigen-combining site having specificity for a different antigen. 
(Hi) Humanized antibodies 

Methods for humanizing non-human antibodies have been described in the art. 
Preferably, a humanized antibody has one or more amino acid residues introduced into 
it from a source which is non-human. These non-human amino acid residues are often 

20 referred to as "import" residues, which are typically taken from an "import" variable 
domain. Humanization can be essentially performed following the method of Winter 
and co-workers (Jones et al, Nature, 321:522-525 (1986); Reichmann et al, 
Nature,332:323-321 (1988); Verhoeyen et al, Science, 239:1534-1536 (1988)), by 
substituting hypervariable region sequences for the corresponding sequences of a 

25 human antibody. Accordingly, such "humanized" antibodies are chimeric antibodies 
(U.S. Patent No. 4,816,567) wherein substantially less than an intact human variable 
domain has been substituted by the corresponding sequence from a non-human 
species. In practice, humanized antibodies are typically human antibodies in which 
some hypervariable region residues and possibly some FR residues are substituted by 

30 residues from analogous sites in rodent antibodies. 

The choice of human variable domains, both light and heavy, to be used in 
making the humanized antibodies is very important to reduce antigenicity. According 



27 



WO 01/97858 PCT/US01/18939 

to the so called "best-fit" method, the sequence of the variable domain of a rodent 
antibody is screened against the entire library of known human variable-domain 
sequences. The human sequence which is closest to that of the rodent is then accepted 
as the human framework region (FR) for the humanized antibody (Suns et al, J. 
5 Immunol, 151:2296 (1993); Chotbia et al, J. Mol Biol, 196:901 (1987)). Another 
method uses a particular framework region derived from the consensus sequence of all 
human antibodies of a particular subgroup of light or heavy chains. The same 
framework may be used for several different humanized antibodies (Carter et al, 
Proc. Natl. Acad. Sci. USA, 89:4285 (1992); Prestae/a/., J. Immunol, 151:2623 
10 (1993)). 

It is further important that antibodies be humanized with retention of high 
affinity for the antigen and other favorable biological properties. To achieve this goal, 
according to a preferred method, humanized antibodies are prepared by a process of 
analysis of the parental sequences and various conceptual humanized products using 

1 5 three-dimensional models of the parental and humanized sequences. Three- 
dimensional immunoglobulin models are commonly available and are familiar to 
those skilled in the art. Computer programs are available which illustrate and display 
probable three-dimensional conformational structures of selected candidate 
immunoglobulin sequences. Inspection of these displays permits analysis of the likely 

20 role of the residues in the functioning of the candidate immunoglobulin sequence, i. 
e., the analysis of residues that influence the ability of the candidate immunoglobulin 
to bind its antigen. In this way, FR residues can be selected and combined from the 
recipient and import sequences so that the desired antibody characteristic, such as 
increased affinity for the target antigen(s), is achieved. In general, the hypervariable 

25 region residues are directly and most substantially involved in influencing antigen 
binding. 

(iv) Ptimatized Antibodies 

Another highly efficient means for generating recombinant antibodies is 
disclosed by Newman, Biotechnology, 10: 1455-1460(1992). More particularly, this 
30 technique results in the generation of primatized antibodies which contain monkey 
variable domains and human constant sequences. This reference is incorporated by 
reference in its entirety herein. Moreover, this technique is also described in 



28 



WO 01/97858 



PCT/US01/18939 



commonly assigned U.S. Application No. 08/379,072, filed on January 25, 1995, 
which is a continuation of U.S. Serial No. 07/912,292, filed July 10, 1992, which is a 
continuation-in-part of U.S. Serial No. 07/856,281, filed March 23, 1992, which is 
finally a continuation-in-part of U.S. Serial No. 07/735,064, filed July 25, 1991. 
5 08/379,072 and the parent application thereof all of which are incorporated by 
reference in their entirety herein. 

This technique modifies antibodies such that they are not antigenically rejected 
upon administration in humans. This technique relies on immunization of 
cynomolgus monkeys with human antigens or receptors. This technique was 
10 developed to create high affinity monoclonal antibodies directed to human cell surface 
antigens. 

Identification of macaque antibodies to human CD20 or CD22 by screening of 
phage display libraries or monkey heterohybridomas obtained using B lymphocytes 
from CD20 or CD22 immunized monkeys can be performed using the methods 

15 described in commonly assigned U.S. Application No. 08/487,550, filed June 7, 1995, 
incorporated by reference in its entirety herein. 

Antibodies generated using the methods described in these applications have 
previously been reported to display human effector function, have reduced 
immunogenicity, and long serum half-life. The technology relies on the fact that 

20 despite the fact that cynomolgus monkeys are phylogenetically similar to humans, 
they still recognize many human proteins as foreign and therefore mount an immune 
response. Moreover, because the cynomolgus monkeys are phylogenetically close to 
humans, the antibodies generated in these monkeys have been discovered to have a 
high degree of amino acid homology to those produced in humans. Indeed, after 

25 sequencing macaque immunoglobulin light and heavy chain variable region genes, it 
was found that the sequence of each gene family was 85-98% homologous to its 
human counterpart (Newman et al., 1992). The first antibody generated in this way, 
an anti-CD4 antibody, was 91-92% homologous to the consensus sequence of human 
immunoglobulin framework regions (Newman et al. , 1 992). 

30 (v) Human antibodies 

As an alternative to humanization, human antibodies can be generated. For 
example, it is now possible to produce transgenic animals (e.g., mice) that are 



29 



WO 01/97858 



PCT/US01/18939 



capable, upon immunization, of producing a full repertoire of human antibodies in the 
absence of endogenous immunoglobulin production. For example, it has been 
described that the homozygous deletion of the antibody heavy-chain joining region 
PH) gene in chimeric and germ-line mutant mice results in complete inhibition of 
5 endogenous antibody production. Transfer of the human germ-line immunoglobulin 
gene array in such germ line mutant mice will result in the production of human 
antibodies upon antigen challenge. See, e.g., Jakobovits et al, Proc. Mad. Acad. Sci. 
USA, 90:255 1 (1993); Jakobovits et al, Nature, 362:255-258 (1993); Bruggermann 
etal, Year in immuno., 7:33 (1993); and US Patent Nos. 5,591,669, 5,589,369 and 
10 5,545,807. 

Alternatively, phage display technology (McCafferty et al, Nature 
348:552-553 (1990)) can be used to produce human antibodies and antibody 
fragments in vitro, from immunoglobulin variable (V) domain gene repertoires from 
unimmunized donors. According to this technique, antibody V domain genes are 

1 5 cloned in-frame into either a major or minor coat protein gene of a filamentous 

bacteriophage, such as Ml 3 or fd, and displayed as functional antibody fragments on 
the surface of the phage particle. Because the filamentous particle contains a 
single-stranded DNA copy of the phage genome, selections based on the functional 
properties of the antibody also result in selection of the gene encoding the antibody 

20 exhibiting those properties. Thus, the phage mimics some of the properties of the B 
cell. Phage display can be performed in a variety of formats; for their review see, e.g., 
Johnson, Kevin S. and Chiswell, David J., Current Opinion in Structural Biology 
3:564-57 1 (1993). Several sources of V-gene segments can be used for phage display. 
Clackson et al, Nature, 352:624-628 (1991) isolated a diverse array of 

25 anti-oxazolone antibodies from a small random combinatorial library of V genes 
derived from the spleens of immunized mice. A repertoire of V genes from 
unimmunized human donors can be constructed and antibodies to a diverse array of 
antigens (including self antigens) can be isolated essentially following the techniques 
described by Marks et al, JMol Biol, 222:581-597 (1991), or Griffith et al, EMBO 

30 J. 12:725-734 (1993). See, also, US Patent Nos. 5,565,332 and 5,573,905. 

Human antibodies may also be generated by in vitro activated B cells (see US 
Patents 20 5,567,610 and 5,229,275). A preferred means of generating human 



30 



WO 01/97858 



PCT/US01/18939 



antibodies using SCID mice is disclosed in commonly-owned, co-pending 
applications. 

(vi) Antibody fragments 

Various techniques have been developed for the production of antibody 
5 fragments. Traditionally, these fragments were derived via proteolytic digestion of 
intact antibodies (see, e.g., Morimoto et al, Journal of Biochemical and Biophysical 
Methods 24:107-1 17 (1992) and Brennan et al, Science, 229:81 (1985)). However, 
these fragments can now be produced directly by recombinant host cells. For example, 
the antibody fragments can be isolated from the antibody phage libraries discussed 

10 above. Alternatively, Fab'-SH fragments can be directly recovered from E. coli and 
chemically coupled to form F(ab')2 fragments (Carter et al, Bio/Technology 10: 
163-167 (1992)). According to another approach, F(ab')2 fragments can be isolated 
directly from recombinant host cell culture. Other techniques for the production of 
antibody fragments will be apparent to the skilled practitioner. In other embodiments, 

15 the antibody of choice is a single chain Fv fragment (scFv). See WO 93/16185; US 
Patent No. 5,571,894; and US Patent No. 5,587,458. The antibody fragment may also 
be a "linear antibody", e.g., as described in US Patent 5,641,870 for example. Such 
linear antibody fragments maybe monospecific or bispecific. 

(vii) Bispecific antibodies 

20 Bispecific antibodies are antibodies that have binding specificities for at least 

two different epitopes. Exemplary bispecific antibodies may bind to two different 
epitopes of the B cell surface marker. Other such antibodies may bind a first B cell 
marker and further bind a second B cell surface marker. Alternatively, an anti-B cell 
marker binding arm may be combined with an arm which binds to a triggering 

25 molecule on a leukocyte such as a T-cell receptor molecule (e.g. CD2 or CD3), or Fc 
receptors for IgG (FcyR), such as FcyRI (CD64), FcyRH (CD32) and FcyRHI (CD 1 6) 
so as to focus cellular defense mechanisms to the B cell. Bispecific antibodies may 
also be used to localize cytotoxic agents to the B cell. These antibodies possess a B 
cell marker-binding arm and an arm which binds the cytotoxic agent (e.g. saporin, 

30 anti-interferon-a, vinca alkaloid, ricin A chain, methotrexate or radioactive isotope 
hapten). Bispecific antibodies can be prepared as full length antibodies or antibody 
fragments (e.g. F(ab)2 bispecific antibodies). 



31 



WO 01/97858 



PCT/US01/18939 



Methods for making bispecific antibodies are known in the art. Traditional 
production of full length bispecific antibodies is based on the coexpression of two 
immunoglobulin heavy chain-light chain pairs, where the two chains have different 
specificities (Millstein et al, Nature, 305:537-539 (1983)). Because of the random 
5 assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) 
produce a potential mixture of 10 different antibody molecules, of which only one has 
the correct bispecific structure. Purification of the correct molecule, which is usually 
done by affinity chromatography steps, is rather cumbersome, and the product yields 
are low. Similar procedures are disclosed in WO 93/08829, and in Traunecker et al, 

10 EMBO J., 10:3655-3659 (1991). 

According to a different approach, antibody variable domains with the desired 
binding specificities (antibody-antigen combining sites) are fused to immunoglobulin 
constant domain sequences. The fusion preferably is with an immunoglobulin heavy 
chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It 

15 is preferred to have the first heavy-chain constant region (CHI) containing the site 
necessary for light chain binding, present in at least one of the fusions. DNAs 
encoding the immunoglobulin heavy chain fusions and, if desired, the 
immunoglobulin light chain, are inserted into separate expression vectors, and are 
co-transfected into a suitable host organism. This provides for great flexibility in 

20 adjusting the mutual proportions of the three polypeptide fragments in embodiments 
when unequal ratios of the three polypeptide chains used in the construction provide 
the optimum yields. It is, however, possible to insert the coding sequences for two or 
all three polypeptide chains in one expression vector when the expression of at least 
two polypeptide chains in equal ratios results in high yields or when the ratios are of 

25 no particular significance. 

In a preferred embodiment of this approach, the bispecific antibodies are 
composed of a hybrid immunoglobulin heavy chain with a first binding specificity in 
one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a 
second binding specificity) in the other arm. It was found that this asymmetric 

30 structure facilitates the separation of the desired bispecific compound from unwanted 
immunoglobulin chain combinations, as the presence of an immunoglobulin light 
chain in only one half of the bispecific molecule provides for a facile way of 



32 



WO 01/97858 PCT/US01/18939 

separation. This approach is disclosed in WO 94/04690. For further details of 
generating bispecific antibodies see, for example, Suresh et ai, Methods in 
Enzymology, 121:210(1986). 

According to another approach described in US Patent No. 5,731,168, the 
5 interface between a pair of antibody molecules can be engineered to maximize the 
percentage of heterodimers which are recovered from recombinant cell culture. The 
preferred interface comprises at least a part of the CH3 domain of an antibody 
constant domain. In this method, one or more small amino acid side chains from the 
interface of the first antibody molecule are replaced with larger side chains (e.g. 
1 0 tyrosine or tryptophan). Compensatory "cavities" of identical or similar size to the 
large side chains) are created on the interface of the second antibody molecule by 
replacing large amino acid side chains with smaller ones (e.g. alanine or threonine). 
This provides a mechanism for increasing the yield of the heterodimer over other 
unwanted end-products such as homodimers. 

15 Bispecific antibodies include cross-linked or "heteroconjugate" antibodies. For 

* . f 

example, one of the antibodies in the heteroconjugate can be coupled to avidin, the 
other to biotin. Such antibodies have, for example, been proposed to target immune 
system cells to unwanted cells (US Patent No. 4,676,980), and for treatment of HTV 
infection (WO 91/00360, WO 92/200373, and EP 03089). Heteroconjugate 

20 antibodies may be made using any convenient cross-linking methods. Suitable 

cross-linking agents are well known in the art, and are disclosed in US Patent No. 
4,676,980, along with a number of cross-linking techniques. 

Techniques for generating bispecific antibodies from antibody fragments have 
also been described in the literature. For example, bispecific antibodies can be 

25 prepared using chemical linkage. Brennan et al, Science, 229:81(1985) describe a 
procedure wherein intact antibodies are proteolytically cleaved to generate F(ab')2 
fragments. These fragments are reduced in the presence of the dithiol complexing 
agent sodium arsenite to stabilize vicinal dithiols and prevent intermolecular disulfide 
formation. The Fab' fragments generated are then converted to thionitrobenzoate 

30 (TNB) derivatives. One of the Fab'-TNB derivatives is then reconverted to the 
Fab'-thiol by reduction with mercaptoethylamine and is mixed with an equimolar 
amount of the other Fab'-TNB derivative to form the bispecific antibody. The 



33 



WO 01/97858 



PCT/USO 1/18939 



bispecific antibodies produced can be used as agents for the selective immobilization 
of enzymes. 

Recent progress has facilitated the direct recovery of Fab'-SH fragments from 
E. coli, which can be chemically coupled to form bispecific antibodies. Shalaby et al, 
5 J.Exp. Med., 175:2 1 7-225 (1 992) describe the production of a fully humanized 
bispecific antibody F(ab') 2 molecule. Each Fab' fragment was separately secreted 
from E. coli and subjected to directed chemical coupling in vitro to form the bispecific 
antibody. The bispecific antibody thus formed was able to bind to cells 
overexpressing the ErbB2 receptor and normal human T cells, as well as trigger the 

1 0 lytic activity of human cytotoxic lymphocytes against human breast tumor targets. 

Various techniques for making and isolating bispecific antibody fragments 
directly from recombinant cell culture have also been described. For example, 
bispecific antibodies have been produced using leucine zippers. Kostelny et al, J. 
Immunol. 148(5):1547-1553 (1992). The leucine zipper peptides from the Fos and Jun 

1 5 proteins were linked to the Fab' portions of two different antibodies by gene fusion. 
The antibody homodimers were reduced at the hinge region to form monomers and 
then re-oxidized to form the antibody heterodimers. This method can also be utilized 
for the production of antibody homodimers. The "diabody" technology described by 
Hollinger et al, Proc.Natl. Acad. Sci. USA, 90:6444-6448 (1993) has provided an 

20 alternative mechanism for making bispecific antibody fragments. The fragments 
comprise a heavy-chain variable domain (Vh) connected to a light-chain variable 
domain (Vijby a linker which is too short to allow pairing between the two domains 
on the same chain. Accordingly, the V H and Vl domains of one fragment are forced to 
pair with the complementary Vl and Vh domains of another fragment, thereby 

25 forming two antigen-binding sites. Another strategy for making bispecific antibody 
fragments by the use of single-chain Fv (sFv) dimers has also been reported. See 
Gruber etal, J. Immunol, 152:5368 (1994). 

Antibodies with more than two valencies are contemplated. For example, 
trispecific antibodies can be prepared. Tutt et al. J. Immunol 147: 60(1991). 

30 Antibody Conjugates and Other Modifications 

The subject therapies may first include the administration of antibody other 
than the radiolabeled CD22 antibody wherein the antibody is attached, e.g. to a 



34 



WO 01/97858 



PCT/US01/18939 



cytotoxin or therapeutic moiety conjugate. 

Chemotherapeutic agents useful in the generation of such antibody-cytotoxic 
agent conjugates have been described above. 

Conjugates of an antibody and one or more small molecule toxins, such as a 
5 calicheamicin, a maytansine (US Patent No. 5,208,020), a trichothene, and CC 1065 
are also contemplated herein. In one preferred embodiment of the invention, the 
antagonist is conjugated to one or more maytansine molecules (e.g. about 1 to about 
1 0 maytansine molecules per antagonist molecule). Maytansine may, for example, be 
converted to May SS-Me which may be reduced to May-SH3 and reacted with 

10 modified antagonist (Charm et al. Cancer Research 52:127-131(1992)) to generate a 
maytansinoid-antagonist conjugate. 

Alternatively, the antibody may be conjugated to one or more caUcheamicin 
molecules. The caUcheamicin family of antibiotics are capable of producing double 
stranded DNA breaks at sub-picomolar concentrations. Structural analogues of 

1 5 calicheamicin which may be used include, but are not limited to, y\, eta, aj, 

N-acetyl-yi 1 , PSAG and 0\ (Hinman et al. Cancer Research 53:3336-3342 (1993) and 
Lode et al, Cancer Research 58: 2925-2928 (1998)). 

Enzymatically active toxins and fragments thereof which can be used include 
diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain 

20 (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, 
alpha sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana 
proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, 
sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, 
enomycin and the tricothecenes. See, for example, WO 93/21232 published October 

25 28,1993. 

The present invention further contemplates antibody conjugated with a 
compound with nucleolytic activity (e.g. a ribonuclease or a DNA endonuclease such 
as a deoxyribonuclease; DNase). 

As discussed above, a variety of radioactive isotopes are available for the 
30 production of radioconjugated antagonists. Examples include At 211 , 1 131 , 1 125 , Y 90 , 
Re 186 , RE 188 , Sm 153 , Bi 212 , P 32 and radioactive isotopes of Lu. 

Conjugates of the antibody and cytotoxic agent may be made using a variety of 



35 



WO 01/97858 PCT7US01/18939 

bifunctional protein coupling agents such as N-succinimidyl-3-(2-pyriyldithiol) 
propionate (SPDP), succinimidyl-4-(N-malehnidomethyl) cyclohexane-l-carboxylate, 
iminotbiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl 
adipunidate HCL), active esters (such as disuccinimidyl suberate), aldehydes (such as 
5 glutareldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), 
bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)-ethylenediamine), 
diisocyanates (such as tolyene 2,6-diisocyanate), and bis-active fluorine compounds 
(such as 1 ,5-difluoro-2, 4-dinitrobenzene). For example, a ricin immunotoxin can be 
prepared as described in Vitetta et al. Science 238: 1098 (1987). Carbon-14-labeled 1 

10 isothiocyanatobenzyl-3- methyldiethylene triaminepentaacetic.acid (MX-DTPA) is an 
exemplary chelating agent for conjugation of radionucleotide to the antagonist. See 
W094/1 1026. The linker may be a "cleavable linker" facilitating release of the 
cytotoxic drug in the cell. For example, an acid-labile linker, peptidase-sensitive 
linker, dimethyl linker or disulfide-containing linker (Charm et al. Cancer Research 

15 52:127-131 (1992)) may be used. 

Alternatively, a fusion protein comprising the antibody and cytotoxic agent 
may be made, e.g. by recombinant techniques or peptide synthesis. 

In yet another embodiment, the antibody may be conjugated to a "receptor" 
(such streptavidin) for utilization in tumor pretargeting wherein the antagonist- 

20 receptor conjugate is administered to the patient, followed by removal of unbound 
conjugate from the circulation using a clearing agent and then administration of a 
"ligand" (e.g. avidin) which is.conjugated to a cytotoxic agent (e.g. a radionucleotide). 

The antibodies of the present invention may also be conjugated with a prodrug 
activating enzyme which converts a prodrug (e.g. a peptidyl chemotherapeutic agent, 

25 see W081/01 145) to an active anti-cancer drug. See, for example, WO 88/07378 and 
U.S. Patent No. 4,975,278. 

The enzyme component of such conjugates includes any enzyme capable of 
acting on a prodrug in such a way so as to covert it into its more active, cytotoxic 
form. 

30 Enzymes that are useful in the method of this invention include, but are not 

limited to, alkaline phosphatase useful for converting phosphate-containing prodrugs 
into free drugs; arylsulfatase useful for converting sulfate-containing prodrugs into 



36 



WO 01/97858 



PCT/US01/18939 



free drugs; cytosine deaminase useful for converting non-toxic5-fluorocytosine into 
the anti-cancer drug, fluorouracil; proteases, such as serratia protease, mermolysin, 
subtilisin, carboxypeptidases and cathepsins (such as cathepsins B and L), that axe 
useful for converting peptide-containing prodrugs into free drugs; 
5 D-alanylcarboxypeptidases, useful for converting prodrugs that contain D-amino acid 
substituents; carbohydratecleaving enzymes such as 13-galactosidase and 
neuraminidase useful for converting glycosylated prodrugs into free drugs; 
13-lactamase useful for converting drugs derivatized with 13-lactams into free drugs; 
and penicillin amidases, such as penicillin V amidase or peniciUin G amidase, useful 

1 0 for converting drugs derivatized at their amine nitrogens with phenoxyacetyl or 
phenylacetyl groups, respectively, into free drugs. Alternatively, antibodies with 
enzymatic activity, also known in the art as "abzymes", can be used to convert the 
prodrugs of the invention into free active drugs (see, e.g., Massey, Nature 
328:457-458 (1987)). Antagonist-abzyme conjugates can be prepared as described 

1 5 herein for delivery of the abzyme to a tumor cell population. 

The enzymes of this invention can be covalently bound to the antagonist by 
techniques well known in the art such as the use of the heterobifunctional crosslinking 
reagents discussed above. Alternatively, fusion proteins comprising at least the 
antigen binding region of an antagonist of the invention linked to at least a 

20 functionally active portion of an enzyme of the invention can be constructed using 
recombinant DNA techniques well known in the art (see, e.g., Neuberger et al, 
Nature, 312:604-608 (1984)). 

Other modifications of the antibody are contemplated herein. For example, the 
antibody may be linked to one of a variety of nonproteinaceous polymers, e.g., 

25 polyethylene glycol, polypropylene glycol, polyoxyalkylenes, or copolymers of 
polyethylene glycol and polypropylene glycol. 

The antibodies disclosed herein may also be formulated as liposomes. 
Liposomes containing the antagonist are prepared by methods known in the art, such 
as described in Epstein et al, Proc. Natl. Acad. Sci. USA, 82:3688 (1985); Hwang et 

30 al, Proc. Natl Acad. Sci. USA, 77:4030 (1980); U.S. Pat. Nos. 4,485,045 and 

4,544,545; and W097/38731 published October 23, 1997. Liposomes with enhanced 
circulation time are disclosed in U.S. Patent No. 5,013,556. 



37 



WO 01/97858 



PCT/US01/18939 



Particularly useful liposomes can be generated by the reverse phase 
evaporation method with a lipid composition comprising phosphatidylcholine, 
cholesterol and PEG derivatized phosphatidylethanolamine (PEG-PE). Liposomes are 
extruded through filters of defined pore size to yield liposomes with the desired 
5 diameter. Fab' fragments of an antibody of the present invention can be conjugated to 
the liposomes as described in Martin et al. J. Biol. Chem. 257:286-288 (1982) via a 
disulfide interchange reaction. A chemotherapeutic agent is optionally contained 
within the liposome. See Gabizon et al. J.National Cancer Inst. 81(19)1484 (1989). 
Amino acid sequence modification(s) of protein or peptide antagonists 

1 0 described herein are contemplated. For example, it may be desirable to improve the 
binding affinity and/or other biological properties of the antibody. Amino acid 
sequence variants of the antibody are prepared by introducing appropriate nucleotide 
changes into the antibody encoding nucleic acid, or by peptide synthesis. Such 
modifications include, for example, deletions from, and/or insertions into and/or 

15 substitutions of, residues within the amino acid sequences of the antagonist. Any 
combination of deletion, insertion, and substitution is made to arrive at the final 
construct, provided that the final construct possesses the desired characteristics. The 
amino acid changes also may alter post-translational processes of the antagonist, such 
as changing the number or position of glycosylation sites. 

20 A useful method for identification of certain residues or regions of the 

antibody that are preferred locations for mutagenesis is called "alanine scanning 
mutagenesis" as described by Cunningham and Wells Science, 244:1081-1085 (1989). 
Here, a residue or group of target residues are identified (e.g., charged residues such 
as arg, asp, his, lys, and glu) and replaced by a neutral or negatively charged amino 

25 acid (most preferably alanine or polyalanine) to affect the interaction of the amino 
acids with antigen. Those amino acid locations demonstrating functional sensitivity to 
the substitutions then are refined by introducing further or other variants at, or for, the 
sites of substitution. Thus, while the site for introducing an amino acid sequence 
variation is predetermined, the nature of the mutation per se need not be 

30 predetermined. For example, to analyze the performance of a mutation at a given site, 
ala scanning or random mutagenesis is conducted at the target codon or region and the 
expressed antagonist variants are screened for the desired activity. 



38 



WO 01/97858 



PCT/US01/18939 



Amino acid sequence insertions include amino- and/or carboxyl-tenninal 
fusions ranging in length from one residue to polypeptides containing a hundred or 
more residues, as well as intrasequence insertions of single or multiple amino acid 
residues. Examples of terminal insertions include an antagonist with an N-terminal 
5 methionyl residue or the antagonist fused to a cytotoxic polypeptide. Other insertional 
variants of the antagonist molecule include the fusion to the N- or C-terminus of the 
antagonist of an enzyme, or a polypeptide which increases the serum half-life of the 
antagonist. 

Another type of variant is an amino acid substitution variant. These variants 
10 have at least one amino acid residue in the antagonist molecule replaced by different 
residue. The sites of greatest interest for substitutional mutagenesis of antibody 
antagonists include the hypervariable regions, but FR alterations are also 
contemplated. Conservative substitutions are shown in Table 1 under the heading of 
"preferred substitutions". If such substitutions result in a change in biological activity, 
15 then more substantial changes, denominated "exemplary substitutions" in Table 1, or 
as further described below in reference to amino acid classes, may be introduced and 
the products screened. 



39 



WO 01/97858 PCT/US01/18939 



Table 1 



Original Residue 


Exemplary Substitutions 


Preferred Substitutions 


Ala (A) 


val; leu; ile 


val 


Arg(R) 


lys; gin; asn 


lys 


Asn(N) . 


gin; his; asp, lys; arg 


gin 


Asp(D) 


glu; asn 


glu 


Cys(C) 


ser; ala 


ser 


Gln(Q 


asn; glu 


asn 


Glu(E) 


asp; gin 


asp 


Gly(G) 


ala 


ala 


ffis(H) 


asn; gin; lys; arg 


arg 


Ile (I) 


Leu; val; met; ala; 
phe; norleucine 


leu 


Leu(L) 


tiorleucine; ile; val; 
met; ala; phe 


ile 


Lys(K) 


arg; gin; asn 


arg 


Met(M) 


leu; phe; ile 


Leu 


Phe(F) 


leu; val; ile; ala; tyr 


tyr 


Pro(P) 


ala 


ala 


Ser(S) 


thr 


thr 


Thr(T) 


ser 


ser 


Trp(W) 


tyr; phe 


tyr 


ryr(Y) 


trp; phe; thr; ser 1 


phe 


Val(V) 


ile; leu; met; phe; 
ala; norleucine 


leu 



Substantial modifications in the biological properties of the antibody are 
accomplished by selecting substitutions that differ significantly in their effect on 
5 maintaining (a) the structure of the polypeptide backbone in the area of the 
substitution, for example, as a sheet or helical conformation, (b) the charge or 
hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain. 
Naturally occurring residues are divided into groups based on common side-chain 
properties: 

10 (1) hydrophobic: norleucine, met, ala, val, leu, ile; 

(2) neutral hydrophiuic: cys, ser, thr; 

(3) acidic: asp, glu; 

(4) basic: asn, gin, his, lys, arg; 

(5) residues that influence chain orientation: gly, pro; and 
15 (6) aromatic: trp, tyr, phe. 



40 



WO 01/97858 PCIYUS01/18939 

Non-conservative substitutions will entail exchanging a member of one of 
these classes for another class. 

Any cysteine residue not involved in maintaining the proper conformation of 
the antagonist also may be substituted, generally with serine, to improve the oxidative 
5 stability of the molecule and prevent aberrant crosslinking. Conversely, cysteine 
bonds) may be added to the antagonist to improve its stability (particularly where the 
antagonist is an antibody fragment such as an Fv fragment). 

A particularly preferred type of substitutional variant involves substituting one 
or more hypervariable region residues of a parent antibody (e.g. a humanized or 

10 human antibody). Generally, the resulting variants selected for further development 
will have improved biological properties relative to the parent antibody from which 
they are generated. A convenient way for generating such substitutional variants is 
affinity maturation using phage display. Briefly, several hypervariable region sites 
(e.g. 6-7 sites) are mutated to generate all possible amino substitutions at each site. 

15 The antibody variants thus generated are displayed in a monovalent fashion from 

filamentous phage particles as fusions to the gene HI product of Ml 3 packaged within 
each particle. The phage-displayed variants are then screened for their biological 
activity (e.g. binding affinity) as herein disclosed. In order to identify candidate 
hypervariable region sites for modification, alanine scanning mutagenesis can be 

20 performed to identified hypervariable region residues contributing significantly to 

antigen binding. Alternatively, or in addition, it may be beneficial to analyze a crystal 
structure of the antigen-antibody complex to identify contact points between the 
antibody and antigen. Such contact residues and neighboring residues are candidates 
for substitution according to the techniques elaborated herein. Once such variants are 

25 generated, the panel of variants is subjected to screening as described herein and 

antibodies with superior properties in one or more relevant assays may be selected for 
further development. 

Another type of amino acid variant of the antibody alters the original 
glycosylation pattern of the antagonist. By altering is meant deleting one or more 

30 carbohydrate moieties found in the antagonist, and/or adding one or more 
glycosylation sites that are not present in the antagonist. 

Glycosylation of polypeptides is typically either N-linked or O-linked. 



41 



WO 01/97858 



PCT/US01/18939 



N-linked refers to the attachment of the carbohydrate moiety to the side chain of an 
asparagine residue. The tripeptide sequences asparagine-X-serine and asparagine- 
X-threonine, where X is any amino acid except proline, are the recognition sequences 
for enzymatic attachment of the carbohydrate moiety to the asparagine side chain. 
5 Thus, the presence of either of these tripeptide sequences in a polypeptide creates a 
potential glycosylation site. O-linked glycosylation refers to the attachment of one of 
the sugars N-aceylgalactosamine, galactose, or xylose to a hydroxyamino acid, most 
commonly seine or threonine, although 5-hydroxyproline or 5-hydroxylysine may also 
be used. 

1 0 Addition of glycosylation sites to the antibody is conveniently accomplished 

by altering the amino acid sequence such that it contains one or more of the 
above-described tripeptide sequences (for N-linked glycosylation sites). The alteration 
may also be made by the addition of, or substitution by, one or more seine or 
threonine residues to the sequence of the original antagonist (for O-linked 

15 glycosylation sites). 

Nucleic acid molecules encoding amino acid sequence variants of the antibody 
are prepared by a variety of methods known in the art. These methods include, but are 
not limited to, isolation from a natural source (in the case of naturally occurring amino 
acid sequence variants) or preparation by oligonucleotide-mediated (or site-directed) 

20 mutagenesis, PCR mutagenesis, and cassette mutagenesis of an earlier prepared 
variant or a non-variant version of the antagonist. 

It may be desirable to modify the antibodies used in the invention to improve 
effector function, e.g. so as to enhance antigen-dependent cell-mediated cyotoxicity 
(ADCC) and/or complement dependent cytotoxicity (CDC) of the antagonist. This 

25 may be achieved by introducing one or more amino acid substitutions in an Fc region 
of an antibody antagonist. Alternatively or additionally, cysteine residue(s) may be 
introduced in the Fc region, thereby allowing interchain disulfide bond formation in 
this region. The homodimeric antibody thus generated may have improved 
intemaUzation capability and/or increased complement- mediated cell killing and 

30 antibody-dependent cellular cytotoxicity (ADCC). See Caron et al, J. Exp Med. 
176:1 191-1 195 (1992) and Shopes, B. J. Immunol. 148:2918-2922 (1992). 
Homodimeric antibodies with enhanced anti-tumor activity may also be prepared 



42 



WO 01/97858 



PCTAJS01/18939 



using heterobifunctional cross-linkers as described in Wolff et al. Cancer Research 
53:2560-2565 (1993). Alternatively, an antibody can be engineered which has dual Fc 
regions and may thereby have enhanced complement lysis and ADCC capabilities. 
See Stevenson et al. Anti-Cancer Drug Design 3:2 19-230 (1989). 
5 To increase the serum half life of the antibody, one may incorporate a salvage 

receptor binding epitope into the antibody (especially an antibody fragment) as 
described in US Patent 5,739,277, for example. As used herein, the term "salvage 
receptor binding epitope" refers to an epitope of the Fc region of an IgG molecule 
(e.g„ IgGI, IgG2, IgG3, or IgG4) that is responsible for increasing the in vivo serum 

1 0 half-life of the IgG molecule. 
Pharmaceutical Formulations 

Therapeutic formulations comprising cold anti-CD20 and radiolabeled anti.- 
CD22 antibodies used in accordance with the present invention are prepared for 
storage by mixing an antagonist having the desired degree of purity with optional 

15 pharmaceutically acceptable carriers, excipients or stabilizers (Remington 's 

Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), in the form of lyophilized 
formulations or aqueous solutions. Acceptable carriers, excipients, or stabilizers are 
nontoxic to recipients at the dosages and concentrations employed, and include 
buffers such as phosphate, citrate, and other organic acids; antioxidants including 

20 ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl 

ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium 
chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl 
paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular 
weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, 

25 gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; 
amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; 
monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, 
or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, 
trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g. 

30 Zn-protein complexes); and/or non-ionic surfactants such as T WEEN™, 
PLURONICS™ or polyethylene glycol (PEG). 

The antibodies may be in the same formulation or may be administered in 



43 



WO 01/97858 



PCT/US01/18939 



difficult formulations. Administration can be concurrent or sequential, and may be 
effective in either order. 

Exemplary anti-CD20 antibody formulations are described in W098/56418, 
expressly incorporated herein by reference. This publication describes a liquid 
5 multidose formulation comprising 40 mg/mL rituximab, 25 mM acetate, 150 mM 
trehalose, 0.9% benzyl alcohol, 0.02% polysorbate 20 at pH 5.0 that has a minimum 
shelf life of two years storage at 2-8°C. Another anti-CD20 formulation of interest 
comprises 1 Omg/mL rituximab in 9.0 mg/mL sodium chloride, 7.35 mg/mL sodium 
citrate dihydrate, 0.7mg/mL polysorbate 80, and Sterile Water for Injection, pH 6.5. 

10 Lyophilized formulations adapted for subcutaneous administration are 

described in W097/04801 Such lyophilized formulations maybe reconstituted with a 
suitable diluent to a high protein concentration and the reconstituted formulation may 
be ad^ninistered subcutaneously to the mammal to be treated herein. 

The formulation herein may also contain more than one active compound as 

1 5 necessary for the particular indication being treated, preferably those with 

complementary activities that do not adversely affect each other. For example, it may 
be desirable to further provide a chemotherapeutic agent, cytokine or 
immunosuppressive agent (e.g. one which acts on T cells, such as cyclosporin or an 
antibody that binds T cells, e.g. one which binds LFA-1). The effective amount of 

20 such other agents depends on the amount of antagonist present in the formulation, the 
type of disease or disorder or treatment, and other factors discussed above. These are 
generally used in the same dosages and with adnunistration routes as used 
hereinbefore or about from 1 to 99% of the heretofore employed dosages. 

The active ingredients may also be entrapped in microcapsules prepared, for 

25 example, by 30 coacervation techniques or by interfacial polymerization, for example, 
hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) 
microcapsules, respectively, in colloidal drug delivery systems (for example, 
liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) 
or in macroemulsions. Such techniques are disclosed in Remington 's Pharmaceutical 

30 Sciences 1 6th edition, Osol, A. Ed. (1 980). 

Sustained-release preparations may be prepared. Suitable examples of 
sustained release preparations include semipermeable matrices of solid hydrophobic 



44 



WO 01/97858 PCT/US01/18939 

polymers containing the antagonist, which matrices are in the form of shaped articles, 
e.g. films, or microcapsules. Examples of sustained-release matrices include 
polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or 
poly(vinylalcohol)), polylactides (U.S. Pat. No. 3,773,919), copolymers of L-glutamic 
5 acid and y ethyl-L-glutamate, noir degradable ethylene-vinyl acetate, degradable lactic 
acid-glycolic acid copolymers such as the LUPRON DEPOT™ (injectable 
microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), 
and poly-D-(-)-3-hydroxybutyric acid. The formulations to be used for in vivo 
adrninistration must be sterile. This is readily accomplished by filtration through 

1 0 sterile filtration membranes. 

Treatment with Cold Anti-CD20 and Hot (Radiolabeled) Anti-CD22 Antibody or 
Antibody Fragment 

A composition comprising cold CD20 antibody, e.g. Rituxan® as a 
radiolabeled anti-CD22 antibody, preferably 90 Y (radiolabeled by use of MXDTPA as 

15 the chelate) will be formulated, dosed, and a<irninistered in a fashion consistent with 
good medical practice. Factors for consideration in this context include the particular 
B cell malignancy, or other condition, e.g. autoimmune, allergy, inflammatory 
disorder, cell therapy or gene therapy, the particular mammal being treated, the 
clinical condition of the individual patient, the cause of the disease or disorder, the 

20 site of delivery of the agent, the method of administration, the scheduling of 

administration, and other factors known to medical practitioners. The therapeutically 
effective amount of the antagonist to be administered will be governed by such 
considerations. 

The CD20 antibody and the radiolabeled CD22 antibody may be in the same 
25 or in different formulations. These formulations can be administered separately or 
concurrently, and in either order. Preferably, the cold CD20 antibody will be 
adtninistered separately from the radiolabeled CD22. 

As a general proposition, the therapeutically effective amount of an antibody 
administered parenterally per dose will typically be in the range of about 0. 1 to 500 
30 mg/kg of patient body weight per day, with the typical initial range of antagonist used 
being in the range of about 2 to 1 00 mg/kg. 

The preferred anti-CD20 antibody is PJTUXAN®. Suitable dosages for such 

45 



WO 01/97858 



PCTYUS01/18939 



antibody are, for example, in the range from about 20mg/m2 to about l'000mg/m2. 
The dosage of the antibody may be the same or different from that presently 
recommended for RITUXAN® for the treatment of non-Hodgkin's lymphoma. For 
example, one may administer to the patient one or more doses of substantially less 
5 than 375mg/m2 of the antibody, e.g. where the dose is in the range from about 

20mg/m 2 to about 250mg/m 2 , for example from about 50mg/m 2 to about 200mg/m 2 . 
The amount of the radiolabeled anti-CD22 antibody will depend upon factors such as 
the particular therapeutic radiolabel, e.g. whether it is an a, P or 8 emitter. Methods 
for determining appropriate dosages of radiation are well known. Preferably, a dosage 

10 will be selected that does not result in myelosuppression severe enough to require one 
marrow or stem cell transplant. 

Preferably, the anti-CD20 antibody will possess substantially B cell deleting 
activity and will induce apoptosis of B cells, comparable to Rituxan®. 

Moreover, one may administer one or more initial doses of the CD20 or 

15 radiolabeled anti-CD22 antibody followed by one or more subsequent dose(s), 

wherein the mg/m 2 dose of the antibody in the subsequent doses) exceeds the mg/m 2 
dose of the antibody in the initial dose(s). For example, the initial dose may be in the 
range from about 20mg/m 2 to about 250mg/m 2 (e.g. from about 50mg/m 2 to about 
200mg/m 2 ) and the subsequent dose maybe in the range from about 250mg/m 2 to 

20 about 1000mg/m 2 . 

As noted above, however, these suggested amounts of both CD20 and CD22 
antibody are subject to a great deal of therapeutic discretion. The key factor in 
selecting an appropriate dose and scheduling is the result obtained, as indicated above. 
For example, relatively higher doses may be needed initially for the treatment of 

25 ongoing and acute diseases. To obtain the most efficacious results, depending on the 
particular B cell malignancy, the antagonist is administered as close to the first sign, 
diagnosis, appearance, or occurrence of the disease or disorder as possible or during 
remissions of the disease or disorder. 

The antibodies are administered by any suitable means, including parenteral, 

30 subcutaneous, intraperitoneal, intrapulmonary, and intranasal, and, if desired for local 
immunosuppressive treatment, intralesional administration. Parenteral infusions 
include intramuscular, intravenous, intraarterial, intraperitoneal, or subcutaneous 



46 



WO 01/97858 



PCT/US01/18939 



administration. In addition, the antibody may suitably be administered by pulse 
infusion, e.g., with declining doses of the antibody. Preferably the dosing is given by 
injections, most preferably intravenous or subcutaneous injections, depending in part 
on whether the a(lministration is brief or chronic. 
5 One additionally may administer other compounds, such as chemotherapeutic 

agents, immunosuppressive agents and/or cytokines with the antibodies herein. The 
combined administration includes co-administration, using separate formulations or a 
single pharmaceutical formulation, and consecutive administration in either order, 
wherein preferably there is a time period while both (or all) active agents 

10 simultaneously exert their biological activities. 

Aside from administration of antibodies to the patient the present application 
contemplates administration of antibodies by gene therapy. Such administration of 
nucleic acid encoding the antibodies is encompassed by the expression "admimstering 
a therapeutically effective amount of an antagonist". See, for example, W096/07321 

15 published March 14, 1996 concerning the use of gene therapy to generate intracellular 
antibodies. 

There are two major approaches to getting the nucleic acid (optionally 
contained in a vector) into the patient's cells; in vivo and ex vivo. For in vivo delivery 
the nucleic acid is injected directly into the patient, usually at the site where the 

20 antagonist is required. For ex vivo treatment, the patient's cells are removed, the 
nucleic acid is introduced into these isolated cells and the modified cells are 
administered to the patient either directly or, for example, encapsulated within porous 
membranes which are implanted into the patient (see, e.g. U.S. Patent Nos. 4,892,538 
and 5,283,187). There are a variety of techniques available for introducing nucleic 

25 acids into viable cells. The techniques vary depending upon whether the nucleic acid 
is transferred into cultured cells in vitro, or in vivo in the cells of the intended host. 
Techniques suitable for the transfer of nucleic acid into mammalian cells in vitro 
include the use of liposomes, electroporation, microinjection, cell fusion, 
DEAF-dextran, the calcium phosphate precipitation method, etc. A commonly used 

30 vector for ex vivo delivery of the gene is a retrovirus. 

The currently preferred in vivo nucleic acid transfer techniques include 
transfection with viral vectors (such as adenovirus, Herpes simplex I virus, or adeno 



47 



WO 01/97858 



PCT/US01/18939 



associated virus) and lipid-based systems (useful lipids for lipid-mediated transfer of 
the gene are DOTMA, DOPE and DC-Choi, for example). In some situations it is 
desirable to provide the nucleic acid source with an agent that targets the target cells, 
such as an antibody specific for a cell surface membrane protein or the target cell, a 
5 ligand for a receptor on the target cell, etc. Where liposomes are employed, proteins 
which bind to a cell surface membrane protein associated with endocytosis may be 
used for targeting and/or to facilitate uptake, e.g. capsid proteins or fragments thereof 
tropic for a particular cell type, antibodies for proteins which undergo internalization 
in cycling, and proteins that target intracellular localization and enhance intracellular 

1 0 half-life. The technique of receptor-mediated endocytosis is described, for example, 
by Wu et al, .1. Biol. Chem 262:4429-4432 (1987); and Wagner et al, Proc. Natl. 
Acad. Sci. USA 87:3410-3414(1990). For review of the currently known gene 
marking and gene therapy protocols see Anderson et al, Science 256:808-8 13 (1992). 
See also WO 93/25673 and the references cited therein. 

15 Articles of Manufacture 

In another embodiment of the invention, an article of manufacture containing 
materials useful for the treatment of the diseases or disorders described above is 
provided. 

The article of manufacture comprises a container and a label or package insert 
20 on or associated with the container. Suitable containers include, for example, bottles, 
vials, syringes, etc. The containers maybe formed from a variety of materials such as 
glass or plastic. The container holds or contains a composition which is effective for 
treating the disease or disorder of choice and may have a sterile access port (for 
example the container may be an intravenous solution bag or a vial having a stopper 
25 pierceable by a hypodermic injection needle). As whole, there may be one or several 
compositions. At least one active agent in one of those compositions is a cold CD20 
antibody, preferably one having substantial B cell depleting activity and at least one 
antibody is therapeutically radiolabeled anti-CD22 antibody or fragment, preferably 
90 Y radiolabeled antibody. The label or package insert indicates that the composition 
30 is used for treating a patient having or predisposed to B cell malignancy, such as those 
listed hereinabove or other conditions or treatment wherein inhibition of B cells is 
desirable, e.g. autoimmune disease, transplant, gene therapy, cell therapy or 



48 



WO 01/97858 



PCT/US01/18939 



inflammatory condition. The article of manufacture may further comprise a second 
container comprising a pharmaceutically acceptable buffer, such as bacteriostatic 
water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose 
solution. It may further include other materials desirable from a commercial and user 
5 standpoint, including other buffers, diluents, filters, needles, and syringes. 

Further details of the invention are illustrated by the following non-limiting 
Examples. The disclosures of all citations in the specification are expressly 
incorporated herein by reference. 

The antibodies of the invention may be administered to a human or other 
10 animal in accordance with the aforementioned methods of treatment in an amount 
sufficient to produce such effect to a therapeutic or prophylactic degree. Such 
antibodies of the invention can be administered to such human or other animal in a 
conventional dosage form prepared by combining the antibody of the invention with a 
conventional pharmaceutically acceptable carrier or diluent according to known 
1 5 techniques. It will be recognized by one of skill in the art that the form and character 
of the pharmaceutically acceptable carrier or diluent is dictated by the amount of 
active ingredient with which it is to be combined, the route of administration and 
other well-known variables. 

The routine of administration of the antibody (or fragment thereof) of the 
20 invention may be oral, parenteral, by inhalation or topical. The term parenteral as 
used herein includes intravenous, intraperitoneal, intramuscular, subcutaneous, rectal 
or vaginal administration. The subcutaneous and intramuscular forms of parenteral 
administration are generally preferred. 

The daily parenteral and oral dosage regimes for employing compounds of the 
25 invention to prophylactically or therapeutically induce immunosuppression, or to 
therapeutically treat carcinogenic tumors will generally be in the range of about 0.05 
to 100, but preferably about 0.5 to 10, milligrams per kilogram body weight per day. 

The antibodies of the invention may also be administered by inhalation. By 
"inhalation" is meant intranasal and oral inhalation administration. Appropriate 
30 dosage forms for such administration, such as an aerosol formulation or a metered 
dose inhaler, may be prepared by conventional techniques. The preferred dosage 



49 



WO 01/97858 



PCT/US01/18939 



amount of a compound of the invention to be employed is generally within the range 
of about 10 to 100 milligrams. 

The antibodies of the invention may also be administered topically. By topical 
administration is meant non-systemic administration and includes the application of 
5 an antibody (or fragment thereof) compound of the invention externally to the 
epidermis, to the buccal cavity and instillation of such an antibody into the ear, eye 
and nose, and where it does not significantly enter the blood stream. By systemic 
administration is meant oral, intravenous, intraperitoneal and intramuscular 
administration. The amount of an antibody required for therapeutic or prophylactic 

10 effect will, of course, vary with the antibody chosen, the nature and severity of the 
condition being treated and the animal. 
Combination Therapy of Invention 

The present invention relates to the combined use of a cold anti-CD20 
antibody and a radiolabeled anti-CD22 antibody targeted to extracellular determinants 

15 of CD20 and CD22. CD20 and CD22 are both antigens present on B-cells. These 
antibodies are selectively reactive under immunological conditions to those 
determinants of CD20 and CD22 displayed on the surface of B-cells and accessible to 
the antibody from the extracellular milieu. 

The term "selectively reactive" or "specific to" includes reference to the 

20 preferential association of an antibody, in whole or part, with a cell or tissue bearing 
the CD22 or CD20 target molecule and not to cells or tissues lacking that target 
molecule. It is, of course, recognized that a certain degree of nonspecific interaction 
may occur between a molecule and a non-target cell or tissue. Nevertheless, specific 
binding, may be distinguished as mediated through specific recognition of the target 

25 CD22 or CD20 molecule. Typically specific binding results in a much stronger 
association between the delivered molecule and cells bearing CD22 or CD20 than 
between the bound molecule and cells lacking CD22 or CD20. Specific binding 
typically results in greater than two-fold, preferably greater than five-fold, more 
preferably greater than ten-fold and most preferably greater than one hundred-fold 

30 increase in amount of bound ligand (per unit time) to a cell or tissue bearing CD22 or 
CD20 as compared to a cell or tissue lacking CD22 or CD20. Specific binding to a 
protein under such conditions requires an antibody that is selected for its specificity 



50 



WO 01/97858 PCT/US01/18939 

for a particular protein. A variety of immunoassay formats are appropriate for 
selecting antibodies specifically immunoreactive with a particular protein. For 
example, solid-phase ELISA immunoassays are routinely used to select monoclonal 
antibodies specifically immunoreactive with a protein. See Harlow and Lane (1 988) 
5 Antibodies, A Laboratory Manual, Cold Spring Harbor Publications, New York, for a 
description of immunoassay formats and conditions that can be used to determine 
specific immunoreactivity. 
Anti-CD20 Antibodies 

The anti-CD20 antibody preferably will comprise a chimeric, humanized or 

10 human monoclonal antibody that specifically bind CD20 or a fragment thereof. Most 
preferably, the anti-CD20 antibody will comprise RITUXAN®, the nucleic acid 
sequence and amino acid sequence of which is reported in U.S. Patent No. 5,736,137, 
incorporated by reference in its entirety herein. This chimeric anti-CD20 antibody 
very effectively depletes B-cells and has been approved for use by the FDA for 

15 treating non-Hodgkin's lymphoma. However, humanized and human monoclonal 
antibodies may also be used. 

The radiolabeled anti-CD22 antibody will preferably comprise a chimeric, 
humanized or human monoclonal antibody or binding fragment thereof specific to 
CD22. In the preferred embodiment, a 90 Y radiolabeled humanized monoclonal 

20 antibody, the sequence of which is disclosed in Leung et al in U.S. Patent 5,789,554, 
issued August 4, 1998, will be utilized. This reference is also incorporated by 
reference in its entirely herein. However, other monoclonal antibodies and binding 
fragments can be substituted therefor. 

Additionally, the use of radionuclides other than 90 Y is contemplated, e.g., 131 I, 

25 67 Cu, 32 P, 125 1, 186 Re, 188 Re, 211 At and the like. Suitable radioisotopes include a, p, 
and y-emitters, Auger electron emitters, and neutron capturing agents that emit a- 
particles or a radioisotype that decays by electron capture. 

The radiolabel may be attached directly or indirectly to the antibody or 
fragment, e.g., by use of a chelating agent. Suitable chelators include by way of 

30 example DTPA and DETA. 



51 



WO 01/97858 



PCT7US01/18939 



Suitable antibody fragments include any antibody which lacks substantially all 
the Fc region of a native antibody. These include in particular scFv, dsFv, Fab, 
FCab 1 )^ F(ab) 2 , Fab, and the like. 

As noted, the anti-CD20 antibody and radiolabeled anti-CD22 antibody can be 
5 administered separately or in combination, and in either order. Preferably, the anti- 
CD20 antibody is administered initially in therapeutically effective amounts, followed 
by the radiolabeled anti-CD22 antibody. 
Binding Affinity of Antibodies 

The antibodies used in this invention are capable of specifically binding an 

1 0 extracellular epitope of CD22 or CD20. An anti-CD22 or anti-CD20 antibody has 
binding affinity for CD22 or CD20 if the antibody binds or is capable of binding 
CD22 or CD20 as measured or determined by standard antibody-antigen assays, for 
example, competitive assays, saturation assays, or standard immunoassays such as 
ELISA or RIA. This definition of specificity applies to single heavy and/or light 

15 chains, CDRS, fusion proteins or fragments of heavy and/or light chains, that are 
specific for CD22 or CD20 if they bind CD22 alone or in combination. 

In competition assays the ability of an antibody to bind a ligand is determined 
by detecting the ability of the antibody to compete with the binding of a compound 
known to bind the ligand. Numerous types of competitive assays are known and are 

20 discussed herein. Alternatively, assays that measure binding of a test compound in the 
absence of an inhibitor may also be used. For instance, the ability of a molecule or 
other compound to bind CD22 can be detected by labeling the molecule of interest 
directly or the molecule be unlabeled and detected indirectly using various sandwich 
assay formats. Numerous types of binding assays such as competitive binding assays 

25 are known (see, e.g., U.S. Patent Nos. 3,376,1 10 and 4,016,043, and Harlow and 
Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Publications, N.Y. 
(1988), which are incorporated herein by reference). Assays for measuring binding of 
a test compound to one component alone rather than using a competition assay are 
also available. For instance, antibodies can be used to identify the presence of the 

30 ligand. Standard procedures for monoclonal antibody assays, such as ELISA, may be 
used (see, Harlow and Lane, supra). For a review of various signal producing systems 



52 



WO 01/97858 



PCT/US01/18939 



which may be used see, U. Patent No. 4,391,904, which is incorporated herein by 
reference. 

The dosages of anti-CD20 to be used in the present invention may vary 
depending on the patient and the antibody used. Chimeric anti-CD20 antibody such 
5 as RITUXIMAB® may be administered at a dosage of at least about 50 mg/m 2 weekly 
for at least 4 weeks. A particularly preferred dosage regimen is about 375 mg/m 2 
weekly for four weeks. 

As noted, preferably after administration of the anti-CD20 antibody, a 
radiolabeled anti-CD22 antibody will be administered, i.e., one which is attached to a 

10 therapeutic radiolabel. Preferred radiolabels are beta emitting isotopes such as 90 Y or 
I31 I, but any radioisotope may be used so long as it may be effectively conjugated to 
the antibody, it has a relatively short decay range, and it succeeds in killing nearby 
cells, i.e., the cells to which it is targeted. Typically, the radiolabel will be attached by 
use of a chelator, e.g., DTPA. 

15 A patient preferably will be treated within one week after administration of the 

depleting anti-CD20 antibody, so long as they are not severely cytopenic, e.g., 
platelets <1 50,000. If the patient is cytopenic following treatment with the depleting 
antibody, recovery should be allowed to occur, e.g. nadir AGC >1000 or platelets 
>1 50,000, before radioimmunotherapy. In cases where cell recovery in the peripheral 

20 blood and/or bone marrow is permitted to occur, more depleting antibody may be 
administered directly before immunotherapy. Such a secondary dosage may be 
administered, for example, at about 250 mg/m 2 for about two weeks directly before or 
overlapping with radioimmunotherapy. 

Dosages of radiolabeled antibodies will also vary depending on the patient, the 
. 25 antibody specificity, half-life, radioisotope stability, etc., and of course, the extent of 
disease. Radiolabeled anti-CD22 antibodies are typically administered at a dosage of 
about 0.001 to 150 mCi/kg, more preferably 0.1 to 50 mCi/kg, still more preferably 
0.1 to 30mCi/kg. Another suitable dosage will range from 10 to 30 mCi/kg. The 
dosages of radiation can be determined by the ordinary artisan. 

30 It should be clear that the treatment methods disclosed herein may be 

combined with other known treatment methods such as chemotherapy or radiotherapy. 
Bone marrow or peripheral blood stem cells may be harvested from said patient 



53 



WO 01/97858 



PCIYUS01/18939 



subsequent to treatment with anti-CD20 antibody and prior to treatment with said 
radiolabeled antibody in order to effect autologous bone marrow or stem cell 
transplantation after radiotherapy. 

It may also be useful to treat patients with cytokines in order to up-regulate the 
5 expression of CD20 or other target protein on the surface of cancerous B cells prior to 
administration of the depleting antibody or the radiolabeled antibody. For up- 
regulation of CD20, cytokines useful for this purpose are EL-4, GM-CSF and TNF- 
alpha. Cytokines may also be administered simultaneously with or prior to or 
subsequent to administration of the depleting antibody or radiolabeled antibody in 

10 order to stimulate immune effector functions. Cytokines useful for this purpose 
include interferon alpha, GM-CSF and G-CSF. 

Chemotherapeutic regimens may be used to supplement the therapies 
disclosed herein, and may be administered simultaneously with or sequentially in any 
order with administration of said radiolabeled antibody. The chemotherapy regimen 

1 5 may be selected from the group consisting of CHOP, ICE, Mitozantrone, Cytarabine, 
DVP, ATRA, Idarubicin, hoelzer chemotherapy regime, La La chemotherapy regime, 
ABVD, CEOP, 2-CdA, FLAG & IDA (with or without subsequent G-CSF treatment), 
VAD, M & P, C-Weekly, ABCM, MOPP and DHAP. A preferred chemotherapeutic 
regimen is CHOP. 

20 The methods of the present invention may be used to treat a variety of B cell 

lymphomas but are particularly useful wherein said B cell lymphoma is non- 
Hodgkin's lymphoma (NHL). RITUXMAB® has already been approved for the 
treatment of low-grade-follicular NHL, but the present inventors have surprisingly 
found that RITUXIMAB® is also beneficial for the treatment of intermediate- and 

25 high-grade NHL, including bulky disease. Accordingly, the lymphomas which are 
treatable by the methods of the present invention include low grade/ follicular non- 
Hodgkin's lymphoma (NHL), small lymphocytic (SL) NHL, intermediate grade/ 
follicular NHL, intermediate grade diffuse NHL, chronic lymphocytic leukemia 
(CLL), high grade immunoblastic NHL, high grade lymphoblastic NHL, high grade 

30 small non-cleaved cell NHL, bulky disease NHL, mantle cell lymphoma, ATDS- 
related lymphoma and Waldenstrom's Macroglobulinemia, so long as such 



54 



WO 01/97858 



PCT/US01/18939 



lymphomas are accompanied by bone marrow involvement which complicates the 
availability of radioimmunotherapy. 

Also, the present invention maybe used to treat autoimmune diseases. 
Examples thereof include glomerulonephritis, Goodpasture's syndrome, necrotizing 
5 vasculitis, lymphadenitis, periarteritis nodosa, systemic lupus erythematosis, arthritis, 
thrombocytopenia purpura, agranulocytosis, autoimmune hemolytic anemias, immune 
reactions against foreign antigens, myasthenia gravis, insulin-resistant diabetes, lupus 
(SLE and drug-induced lupus). Further, the present invention may be used to treat or 
prevent humoral immune responses against transplanted cells, tissues or organs. 
1 0 Exemplary treatment conditions will now be illustrated by the following. 

EXAMPLE 1 

A patient with non-Hodgkin's is initially treated with RITUXAN®. This 
initial treatment comprises administration of 375 mg/m 2 RITUXAN® weekly for four 
15 weeks. 

A week after this RITUXAN® antibody regimen is completed, the patient is 
then treated with a 90 Y radiolabeled humanized anti-CD22 antibody (humanized LL2 
antibody disclosed in U.S. Patent 5,789,554, to Leung et al, assigned to 
hnmunomedics, incorporated by reference in its entire herein.) The patient is treated 
20 with a dosage of ^Y-labeled anti-CD22 antibody ranging from 1 0 to 3 0 mCi. 

EXAMPLE 2 

A patient who is to be transplanted with a kidney is treated prior to transplant 
with RITUXAN® at a dosage of 375 mg/m 2 weekly for four weeks in order to deplete 
B-cells prior to transplant and thereby reduce the likelihood of a humoral immune 
25 response against the transplanted organ. 

Concurrent or within a week after RITUXAN® treatment, the subject is 
treated with low dosage of 90 Y radiolabeled humanized anti-CD22 monoclonal 
antibody, i.e., at a dosage of 10 to 30 mCi. The treated subject is then transplanted 
with the kidney by conventional surgical methods. Preferably, the subject will also be 
30 treated with anti-CD40L, anti-B7 or other immunosuppressants, e.g., cyclosporin. 



55 



WO 01/97858 



PCT/US01/18939 



WHAT IS CLAIMED IS: 

1 . A method of treating a disease or condition wherein suppression and/or 
depletion and/or blocking the function of B-cells is therapeutically beneficial, 

5 comprising the steps of: 

(i) administering a therapeutically effective amount of cold (non- 
radiolabeled) anti-CD20 monoclonal antibody having B cell depleting 
activity substantially equivalent to Rituxan®; and 

(ii) a<toinistering a therapeutically effective amount of a hot (radiolabeled) 
10 anti-CD22 antibody or fragment thereof; 

wherein said anti-CD20 antibody and said radiolabeled anti-CD22 antibody or 
fragment are administered separately or in combination, and in either order. 

2. The method of Claim 1, which is used to treat a B-cell malignancy, 
15 leukemia or lymphoma. 

3. The method of Claim 1, which is used to treat an autoimmune disease. 

4. The method of Claim 2, wherein said disease is non-Hodgkins 
20 lymphoma. 

5. The method of Claim 2, wherein said malignancy, leukemia or 
lymphoma is selected from the group consisting of low grade/ follicular non- 
Hodgkin's lymphoma (NHL), small lymphocytic (SL) NHL, intermediate grade/ 

25 follicular NHL, intermediate grade diffuse NHL, chronic lymphocytic leukemia 

(CLL), high grade immunoblastic NHL, high grade lymphoblastic NHL, high grade 
small noncleaved cell NHL, bulky disease NHL, mantle cell lymphoma, AIDS-related 
lymphoma and Waldenstrom's Macroglobulinemia. 

30 6. The method of Claim 1, wherein the amount of said cold anti-CD20 

antibody ranges from 0.1 mg to 500 mg/m 2 per week. 



56 



WO 01/97858 



PCT/US01/18939 



7. The method of Claim 6, wherein the amount of said cold anti-CD20 
antibody is at least about 500 mg/m 2 weekly. 

8. The method of Claim 7, wherein said dosage is about 375 mg/m 2 
5 weekly for four weeks. 

9. The method of Claim 1 , wherein said radiolabeled anti-CD22 antibody 
or fragment is an yttrium-labeled anti-CD22 antibody or fragment. 

10 10. The method of Claim 6, wherein said radiolabeled anti-CD22 antibody 

fragment is a Fab 2 , Fab, Fv, or domain deleted antibody. 

1 1 . The method of Claim 9, wherein said anti-CD22 antibody is a 90 Y 
labeled humanized LL2 antibody. 

15 

12. The method of Claim 11, wherein the dosage of said radiolabeled 
antibody ranges from 10 to 30 mCi. 

1 3 . The method of Claim 1 2, wherein said radiolabeled anti-CD22 

20 antibody is administered about a week after an RITUXAN® antibody therapeutic 
regimen has been completed. 

14. The method of Claim 13, which further includes administration of a 
chemotherapeutic agent. 

25 

15. The method of Claim 13, which further includes administration of a 
cytokine. 

16. The method of Claim 1 wherein the anti-CD20 antibody is Rituxan®. 

30 

17. The method of Claim 1 6 wherein the condition treated is a B cell 
malignancy, leukemia or lymphoma. 



57 



WO 01/97858 



PCT/US01/18939 



18. The method of Claim 5 wherein the anti-CD20 antibody is Rituxan®. 

19. The method of Claim 1 6 wherein the disease is transplant. 

5 

20. The method of Claim 1 6 wherein the condition is cell therapy. 

2 1 . The method of Claim 1 6 wherein the disease is an autoimmune 
disease. 

10 

22. The method of Claim 2 1 wherein the autoimmune disease is a B cell 
related autoimmune disease. 

23 . The method of Claim 1 wherein the condition is cell therapy. 

15 

24. The method of Claim 1 wherein the condition is allergy. 

25. The method of Claim 1 wherein the condition is a treatment involving 
the administration of an antigenic moiety. 

20 

26. The method of Claim 25 wherein the antigenic moiety is a therapeutic 
protein. 

27. The method of Claim 1 wherein the disease is a solid tumor wherein B 
25 cells promote tumor growth but are not themselves the origin of the tumor. 



58 



(12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) 



(19) World Intellectual Property Organization 
International Bureau 




; IIIIIJ'II I Hi'lll.lM M l.li;] 1MIIIIIII llli.llll. 11,11; llll.hllfl 



(43) International Publication Date (10) International Publication Number 

27 December 2001 (27.12.2001) PCT WO 01/097858 A3 



(51) International Patent Classification 7 : A6IK 51/10, 
47/48, A6 IP 35/00 

(21) International Application Number: PCT/US01/18939 

(22) International Filing Date: 14 June 2001 (14.06.2001) 

(25) Filing Language: English 

(26) Publication Language: English 



(30) Priority Data: 

60/212,668 



20 June 2000 (20.06.2000) US 



(71) Applicant: IDEC PHARMACEUTICALS CORPORA- 
TION [US/US]; 3030 Callan Road, San Diego, CA 92121 
(US). 

(72) Inventor: WHITE, Christine; P.O. Box 9242, Rancho 
Santa Fe, CA 92067 (US). 

(74) Agents: TESKIN, Robin, L. et ai.; Pillsbury Winthrop 
LLP, 1600 Tysons Boulevard, McLean, VA 22102 (US). 

(81) Designated States (national): AE, AG, AL, AM, AT, AU, 
AZ, BA, BB, BG, BR, BY, BZ, CA, CH, CN, CO, CR, CU, 



CZ, DE, DK, DM, DZ, EC, EE, ES, FI, GB, GD, GE, GH, 
GM, MR, HU, ID, IL, IN, IS, JP, KE, KG, KP, KR, KZ, LC, 
LK, LR, LS, LT, LU, LV, MA, MD, MG, MK, MN, MW, 
MX, MZ, NO, NZ, PL, PT, RO, RU, SD, SE, SG, SI, SK, 
SL, TJ, TM, TO, TT, TZ, UA, UG, UZ, VN, YU, ZA, ZW. 

(84) Designated States (regional): ARIPO patent (GH, GM, 
KE, LS, MW, MZ, SD, SL, SZ, TZ, UG, ZW), Eurasian 
patent (AM, AZ, BY, KG, KZ, MD, RU, TJ, TM), European 
patent (AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, 
IT, LU, MC, NL, PT, SE, TR), OAPI patent (BF, BJ, CF, 
CG, CI, CM, GA, GN, GW, ML, MR, NE, SN, TD, TG). 

Published: 

— with international search report 

— before the expiration of the time limit for amending the 
claims and to be republished in the event of receipt of 
amendments 

(88) Date of publication of the international search report: 

8 August 2002 

For two-letter codes and other abbreviations, refer to the "Guid- 
ance Notes on Codes and Abbreviations" appearing at the begin- 
ning of each regular issue of the PCT Gazette. 



< 

00 
ITS 
00 



^ (54) Title: COLD ANTI-CD20 ANTIBODY/RADIOLABELED ANTI-CD22 ANTIBODY COMBINATION 



(57) Abstract: Treatment of B-cell associated diseases including autoimmune and B-cell malignancies such as leukemias, lym- 
phomas, using the combination of an anti-CD20 antibody, preferably R1TUXAN® and a radiolabeled anti-CD22 antibody, prefer- 
ably an '"'Y labeled humanized anti-CD22 antibody, is described. These therapeutic regimens provide for enhanced depletion of B 
cells, and therefore reduce the risk in B cell malignancy treatment of relapse associated with RTTUXAN 11 ' and, moreover, provide 
for prolonged immunosuppression of B-cell immune responses, especially in the context of autoimmune diseases and transplant. 



INTERNATIONAL SEARCH REPORT 



In" 1 " lonal Application No 

PUT/US 01/18939 



A. CLASSIFICATION OF SUBJECT MATTER 

IPC 7 A61K51/10 A61K47/48 A61P35/00 



According to International Patent Classification (IPC) or to bolti national classification and IPC 



B. FIELDS SEARCHED 



Minimum documentation searched (classification system Mowed by classification symbols) 

IPC 7 A61K 



Documentation searched other than minimum documentation to the extent that such documents are included In the fields searched 



Electronic data base consulted during the international search (name of data base and, where practical, search terms used) 

CHEM ABS Data, EMBASE , BIOSIS, EPO-Internal 



C. DOCUMENTS CONSIDERED TO BE RELEVANT 



Category • Citation of document, with Indication, where appropriate, of the relevant passages 



Relevant to claim No. 



WO 98 42378 A (GOLDENBERG DAVID M 
;IMMUN0MEDICS INC (US)) 
1 October 1998 (1998-10-01) 
cited in the application 
claims 1,9,14,15,18 

US 5 789 554 A (HANSEN HANS ET AL) 

4 August 1998 (1998-08-04) 

cited 1n the application 

claims; example 9 

WO 00 20864 A (BIOCRYSTAL LTD) 
13 April 2000 (2000-04-13) 
cited 1n the application 

_/-- 



1-27 



1-27 



X Further documents are listed in the continuation of box C. 



El 



Patent family members are listed In annex. 



° Special categories of cited documents : 

•A" document defining the general slate of the art which Is not 
considered to be of particular relevance 

'E' earlier document but published on or after the international 
filing date 

*L" document which may throw doubts on priority claim(s)or 
which is cited to establish the publication date of another 
citation or other special reason (as specified) 

"O" document referring to an oral disclosure, use, exhibition or 
other means 

"P" document published prior to the international filing date but 
later than the priority date claimed 



"T" later document published after the International filing date 
or priority date and not in conflict with the application but 
cited to understand the principle or theory underlying the 
invention 

'X' document of particular relevance; the claimed Invention 
cannot be considered novel or cannot be considered to 
involve an inventive step when the document Is taken alone 

■Y" document of particular relevance; the claimed invention 
cannot be considered to Involve an inventive step when the 
document Is combined with one or more other such docu- 
ments, such combination being obvious to a person skilled 
In the art. 

'&• document member of the same patent family 



Dale of the actual completion of the international search 



24 May 2002 



Date of mailing of the international search report 



03/06/2002 



Name and mailing address of the ISA 

European Patent Office, P.B. 5816 Patentlaan 2 
NL-2280HVRIjSWijk 
TeL (+31-70) 340-2040, Tx. 31 651 epo nl, 
Fax: (+31-70) 340-3016 



Authorized officer 



Berte, M 



Foim PCT/ISA/210 (second sheet) (July 1892) 



page 1 of 2 



INTERNATIONAL SEARCH REPORT 


lnf~ "onal Application No 

PCT/US 01/18939 


C(Contlnuatlon) DOCUMENTS CONSIDERED TO BE RELEVANT 


Category • 


Citation of document, with Indlcation.where appropriate, of the relevant passages 


Relevant lo claim No. 


p,x 


WO 00 74718 A (HANSEN HANS J ;GOLDENBERG 
DAVID M (US); IMMUNOMEDICS INC (US)) 
14 December 2000 (2000-12-14) 
claims 1 , 12-18,21 , 31 




1-27 


P,Y 


WO 01 10461 A (RASTETTER WILLIAM ;WHITE 
CHRISTINE (US); IDEC PHARMA CORP (US)) 
15 February 2001 (2001-02-15) 
claims 




1-27 


P,A 


WO 00 67796 A (GENENTECH INC ; IDEC 
PHARMACEUTICALS INC (US)) 
16 November 2000 (2000-11-16) 
claims 1,2,9,11 




1-27 


E 


WO 02 22212 A (IDEC PHARMA CORP) 
21 March 2002 (2002-03-21) 
claims 1-8 




1 


E 


W0 02 04021 A (IDEC PHARMA CORP) 
17 January 2002 (2002-01-17) 
claims 1-13 




1 


P,X 


US 6 090 365 A (BUTCHKO GREGORY M ET AL) 
18 July 2000 (2000-07-18) 
claims 1,11,13 




1-27 



Form PCT/ISA/210 (continuation of second sheet) (July 1992) 



page 2 of 2 



IN 1 bKIMA 1 ICJNAL SEARCH REPORT 

Information on patent family members 


lj tlonal Application No 

PTJT/US 01/18939 


Patent document 
cited in search report 


Publication 
date 


Patent family 
member(s) 


Publication 
date 



WO 


9842378 


A 


01- 


•10- 


-1998 


AU 


728325 B2 


04-01-2001 














All 

AU 


o/oiuy© a 


on in iciqo 
tU-lU-iyyo 














TP 
tr 


OOAQfiAA Al 

uyoyooo mi 


1 9— 01 -9noo 














IP 
Or 


9001 kibosh t 


10— 1U— tUU 1 














Uo 


DlOO/Hf 01 


oa- 09_9noi 
















QQ/1927Q Al 


u 1— 1U— iyro 














UO 


O0UOJ7J Ol 


0-9001 
















7A 


JOUL'f JO M 


04-1 1-1 QQA. 


US 


5789554 


A 


04- 


-08- 


-1998 


US 


6187287 Bl 


13-02-2001 














All 


1979fiOi; A 


u/ uj iyyo 














PA 




99— (19— 1 QQA 

tt Ut 133U 














TP 

tr 


(1771 90S A1 
U/ / l&Uo HI 


07— OK— 1 QQ7 

u/ uo— iyy / 














Tl 

1L 


li'jyuy h 


9Q— 1 0—1 QQQ 

to-iu-iyyy 














JP 


3053873 B2 


19-06-2000 














JP 


10505231 T 


26-05-1998 
















yoO'ly^b Al 


22-02-lyyo 


wo 


0020864 


A 


13- 


-04- 


-2000 


US 


6224866 Bl 


01-05-2001 














AU 


1442700 A 


26-04-2000 














Er 


1 t not a At 

1127260 Al 


OA AO AAA1 

29-08-2001 














1 IA 

WO 


0020864 Al 


1 0 Ail onnn 

13-04-2000 














US 


2001033839 Al 


25-10-2001 


wo 


0074718 


A 


14- 


-12- 


-2000 


AU 


5600500 A 


28-12-2000 














EP 


1194167 Al 


10-04-2002 














WO 


0074718 Al 


14-12-2000 


wo 


0110461 


A 


15- 


-02- 


-2001 


AU 


7052100 A 


05-03-2001 














WO 


0110461 Al 


15-02-2001 



WO 


0067796 


A 


16- 


-11- 


-2000 


AU 


4714300 A 


21-11-2000 














BR 


0011197 A 


19-02-2002 














EP 


1176981 Al 


06-02-2002 














NO 


20015417 A 


07-01-2002 














WO 


0067796 Al 


16-11-2000 


WO 


0222212 


A 


21- 


-03- 


-2002 


WO 


0222212 A2 


21-03-2002 














WO 


0222687 A2 


21-03-2002 


WO 


0204021 


A 


17- 


-01- 


-2002 


US 


2002006404 Al 


17-01-2002 














WO 


0204021 Al 


17-01-2002 














US 


2002028178 Al 


07-03-2002 



US 6090365 


A 


18-07-2000 US 


5595721 A 


21-01-1997 






US 


6287537 Bl 


11-09-2001 






US 


5843398 A 


01-12-1998 






US 


6015542 A 


18-01-2000 



Form PCT/ISA/210 (patent lamlly annex) (July 1892)